WO2014100323A1 - Polycyclic-carbamoylpyridone compounds and their pharmaceutical use - Google Patents

Polycyclic-carbamoylpyridone compounds and their pharmaceutical use Download PDF

Info

Publication number
WO2014100323A1
WO2014100323A1 PCT/US2013/076367 US2013076367W WO2014100323A1 WO 2014100323 A1 WO2014100323 A1 WO 2014100323A1 US 2013076367 W US2013076367 W US 2013076367W WO 2014100323 A1 WO2014100323 A1 WO 2014100323A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
mmol
hiv
hydrogen
reaction mixture
Prior art date
Application number
PCT/US2013/076367
Other languages
French (fr)
Inventor
Haolun Jin
Scott E. Lazerwith
Teresa Alejandra Trejo Martin
Elizabeth M. Bacon
Jeromy J. Cottell
Zhenhong R. Cai
Hyung-Jung Pyun
Philip Anthony Morganelli
Mingzhe Ji
James G. TAYLOR
Xiaowu Chen
Michael R. Mish
Manoj C. Desai
Original Assignee
Gilead Sciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=49917297&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2014100323(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Gilead Sciences, Inc. filed Critical Gilead Sciences, Inc.
Priority to KR1020237014240A priority Critical patent/KR20230061576A/en
Priority to KR1020157019194A priority patent/KR101770048B1/en
Priority to PH12019501848A priority patent/PH12019501848A1/en
Priority to BR122015029881-4A priority patent/BR122015029881B1/en
Priority to KR1020217012463A priority patent/KR102351912B1/en
Priority to NZ709260A priority patent/NZ709260A/en
Priority to KR1020207015960A priority patent/KR102246963B1/en
Priority to DK13815937.1T priority patent/DK2822954T3/en
Priority to EA201591027A priority patent/EA030003B1/en
Priority to EP22178867.2A priority patent/EP4122935A1/en
Priority to UAA201506209A priority patent/UA114351C2/en
Priority to EP15201519.4A priority patent/EP3067358B1/en
Priority to MX2017000362A priority patent/MX357940B/en
Priority to CN201380073134.XA priority patent/CN104995198B/en
Priority to MDA20200093A priority patent/MD4841B1/en
Priority to MEP-2016-93A priority patent/ME02400B/en
Priority to MDA20170067A priority patent/MD4754C1/en
Priority to KR1020227018650A priority patent/KR102527797B1/en
Priority to CA2893843A priority patent/CA2893843C/en
Priority to RS20160407A priority patent/RS54873B1/en
Priority to BR112015014714-3A priority patent/BR112015014714B1/en
Priority to JP2015549669A priority patent/JP6028105B2/en
Priority to CR20170279A priority patent/CR20170279A/en
Priority to KR1020177016697A priority patent/KR102040007B1/en
Priority to SI201330197A priority patent/SI2822954T1/en
Priority to CN202310322212.0A priority patent/CN116640140A/en
Priority to MDA20150064A priority patent/MD4736C1/en
Priority to KR1020197031864A priority patent/KR102120875B1/en
Priority to KR1020227001078A priority patent/KR102406288B1/en
Priority to ES13815937.1T priority patent/ES2577283T3/en
Priority to PL15201519T priority patent/PL3067358T3/en
Priority to CN202010188107.9A priority patent/CN111303152B/en
Priority to EP13815937.1A priority patent/EP2822954B1/en
Priority to MX2015008009A priority patent/MX344879B/en
Priority to EP19185121.1A priority patent/EP3608325B1/en
Priority to SG11201504857SA priority patent/SG11201504857SA/en
Priority to AU2013361401A priority patent/AU2013361401C1/en
Publication of WO2014100323A1 publication Critical patent/WO2014100323A1/en
Priority to IL239316A priority patent/IL239316A/en
Priority to PH12015501445A priority patent/PH12015501445B1/en
Priority to HK15105909.2A priority patent/HK1205124A1/en
Priority to CR20150380A priority patent/CR20150380A/en
Priority to ZA2015/07997A priority patent/ZA201507997B/en
Priority to PH12016500389A priority patent/PH12016500389B1/en
Priority to HK16104629.3A priority patent/HK1216643A1/en
Priority to HRP20160544TT priority patent/HRP20160544T1/en
Priority to IL245780A priority patent/IL245780B/en
Priority to SM201600157T priority patent/SMT201600157B/en
Priority to IL247311A priority patent/IL247311A0/en
Priority to AU2016262722A priority patent/AU2016262722B2/en
Priority to LU00083C priority patent/LUC00083I2/en
Priority to LTPA2018511C priority patent/LTC2822954I2/en
Priority to NL300947C priority patent/NL300947I2/en
Priority to CY2018022C priority patent/CY2018022I2/en
Priority to HUS1800035C priority patent/HUS1800035I1/en
Priority to AU2018232957A priority patent/AU2018232957B2/en
Priority to CY20191101151T priority patent/CY1122246T1/en
Priority to AU2020202914A priority patent/AU2020202914B2/en
Priority to IL274988A priority patent/IL274988A/en
Priority to AU2021221427A priority patent/AU2021221427B2/en
Priority to AU2023286038A priority patent/AU2023286038A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/529Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/537Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines spiro-condensed or forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/18Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/22Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed systems contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • HIV human immunodeficiency vims
  • Human immunodeficiency virus infection and related diseases are a major public health problem worldwide.
  • Human immunodeficiency vims type 1 (HTV-1) encodes three enzymes which are required for viral replication: reverse transcriptase, protease, and integrase.
  • reverse transcriptase a enzyme which is required for viral replication
  • protease a enzyme which is required for viral replication
  • integrase integrase.
  • drugs targeting reverse transcriptase and protease are in wide use and have shown effectiveness, particularly when employed in combination, toxicity and development of resistant strains have limited their usefulness (Palella, et al. N. Engl. J Med, (1998) 338:853 -860; Richman, D. D. Nature (2001) 410:995-1001).
  • PXR Pregnane X receptor
  • Activation of PXR is known to up-regulate or induce the production of metabolic enzymes such as cytochrome P450 3A4 (CYP3A4) as well as enzymes involved in transport, such as OATP2 in the liver and intestine (Endocrine Reviews (2002) 23(5): ⁇ 87-702).
  • CYP3A4 cytochrome P450 3A4
  • OATP2 oxygen species
  • PXR activation it is desirable to minimize PXR activation. Further, it is known that PXR is activated by many different classes of molecules [Endocrine Reviews (2002) 23(5):687-702). Thus for drugs that will be co-administered with other drugs, it is important to test for and minimize PXR activation.
  • Transporters have been identified as playing a role in the pharmacokinetic, safety and efficacy profile or drugs, and certain drug-drug interactions are mediated by transporters. See, Giacomini KM, et al. ""Membrane transporters in drug development,” NaLRev Drug Discov. 9: 215-236, 2010; Zhang L, et al. "Transporter- Mediated Drug-Drug Interactions,” Clin. Pharm. Ther. 89(4):481-484 (2011).
  • OCT2 organic cation transporter 2
  • SLC22A2 solute carrier
  • MATE multidrug and toxin extrusion
  • Inhibition of OCT2 can thus lead to increased levels of serum creatinine and the potential for increased levels of other OCT2 substrates, it is important as well to test and reduce OCT2 inhibition of drugs.
  • a goal of antiretroviral therapy is to achieve viral suppression in the HIV infected patient.
  • Treatment guidelines published by the United States Department of Health and Human Services provide that achievement of viral suppression requires the use of combination therapies, i.e., several drugs from at least two or more dr g classes.
  • combination therapies i.e., several drugs from at least two or more dr g classes.
  • the present invention is directed to novel polycyclic carbamoylpyridone compounds, having antiviral activity, including stereoisomers and pharmaceutical [y acceptable salts thereof, and the use of such compounds in the treatment of HIV infections.
  • the compounds of the invention may be used to mhibit the activity of HIV integrase and may be used to reduce HIV replication.
  • X is ⁇ () ⁇ or -NZ 3 - or -CHZ 3 -;
  • W is -CHZ 2 -;
  • Z 1 , Z z and Z 3 are each, independently, hydrogen or Cj -3 alkyl, or wherein Z 1 and Z 2 or Z 1 and Z 3 , taken together, form -L- wherein L is -C(R a ) 2 -, -C(R a ) 2 C(R a ) 2 -, -C(R a ) 2 C(R a ) 2 C(R a ) 2 -, or -C(R a ) 2 C(R a ) 2 C(R a ) 2 C(R a ) 2 -, wherein at least one of Z 1 and Z 2 or Z x and Z 3 , taken together, form -L-; Z is a bond, -CH 2 -, or ⁇ ( !! ( I !> ⁇ ;
  • Y 1 and Y 2 are each, independently, hydrogen, Cj-aalkyl or R 3 ⁇ 4 is phenyl substituted with one to three halogens;
  • each R 3 is, independently, hydrogen, halo, hydroxyl or ( ' ⁇ salky].
  • X is -O- or -NZ 3 - or -CHZ 3 -;
  • W is -O- or -NZ 2 - or -CHZ 2 -:
  • Z 1 , Z 2 and Z 3 are each, independently, hydrogen or C 1-3 alkyl, or wherein Z 1 and Z 2 or Z l and Z 3 , taken together, form -L- wherein L is -C(R a ) 2 -, -C(R a ) 2 C(R a ) 2 -, -C(R a ) 2 C(R a ) 2 C(R a )2-, -C(R a ) 2 C(R a ) 2 C(R a ) 2 C(R a )2-, -C(R a ) 2 OC(R a ) 2 -,
  • Z is a bond or -CH 2 -, ⁇ ( ' !! . ⁇ ( H . -. -CH 2 CH 2 CH 2 -, -CH 2 OCH 2 -, -CH 2 NR a CH 2 -, -CH 2 SCH 2 -,-CH 2 S(0)CH 2 - or -CH 2 S0 2 CH 2 -;
  • Y 1 and Y i are each, independently, hydrogen, or Ci.3h.al0al.kyl, or Y 1 and Y 2 , together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms, wherein the carbocyciic or heterocyclic ring is optionally substituted with one or more R ;
  • R 5 is optionally substituted aryl or optional!)' substituted heteroaryl; and each R a is, independently, hydrogen, halo, hydroxyl or C 1 . 4 alkyl, or wherein two R a groups, together with the carbon atom to which they are attached, form
  • a pharmaceutical composition comprising a compound having Formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the invention also provides the use of a pharmaceutical composition as described hereinabove for the treatment of an HIV infection in a human being having or at risk of having the infection.
  • a method of using a compound having Formula (I) in therapy comprising administering to the mammal a compound having Formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
  • an article of manufacture comprising a composition effective to treat an HIV infection; and packaging material comprising a label which indicates that the composition can be used to treat infection by HIV is disclosed.
  • exemplary compositions comprise a compound of Formula (I) according to this invention or a pharmaceutically acceptable salt thereof.
  • a method of inhibiting the replication of HIV comprises exposing the virus to an effective amount of the compound of Formula (I), or a salt thereof, under conditions where replication of HIV is inhibited.
  • a compound of Formula (I) or “compounds of Formula (I)” refers to ail embodiments of Formula (I), including, for example, compounds of Formulas (II-A), (II-B), (II-C), (III-A), ( ⁇ - ⁇ ), (III-C), (III-D), (II -E), (III-F), (ITI-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (TV-AD), (IV-AE), (IV-AF), (TV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as well as the specific compounds disclosed herein.
  • Amino refers to the -N3 ⁇ 4 radical.
  • Niro refers to the -N0 2 radical.
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated ⁇ i.e., contains one or more double and/or triple bonds), having from one to twelve carbon atoms (Ci-Cj?
  • alkyl preferably one to eight carbon atoms (Cj-Cg alkyl) or one to six carbon atoms (Ci-Ce alkyl), and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, H-propyl, l-rnethylethyl (zso-propyl), «-butyl, n-pentyl, 1 ,1-dirnethylethyl (/-butyl), 3-methylhexyl, 2-meth.ylh.exyi, etherryl, prop-l-enyl, but-l -enyl, pent-l-enyl, penta-l.,4-dienyl, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like.
  • an alkyl group may be optionally substituted
  • Alkylene or “alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, n-butylene, ethenylene, propenylene, n-butenylene, propynylene, « ⁇ butynylene, and the like.
  • the alkylene chain is attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond.
  • the points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkylene chain may be optionally substituted.
  • Alkoxy refers to a radical of the formula -ORA where R A is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxy group may be optionally substituted.
  • Alkylamino refers to a radical of the formula -NHR A or -NR A RA where each is, independently, an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylamino group may be optionally substituted.
  • Thioalkyl refers to a radical of the formula -SRA where A is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a thioalkyl group may be optionally substituted.
  • Aryl refers to a monocylic hydrocarbon ring system radical comprising hydrogen and 6 to 18 carbon atoms.
  • Aryl radicals include, but are not limited to, aryl radicals derived from benzene.
  • aryl or the prefix “ar-” (such as in “aralkyl”) is meant to include aryl radicals that are optionally substituted.
  • Araikyi refers to a radical of the formula -R B -RC where R B is an alkylene chain as defined above and c is one or more aryl radicals as defined above, for example, benzyl. Unless stated otherwise specifically in the specification, an aralkyl group may be optionally substituted.
  • Cycloalkyl or “carbocyclic ring” refers to a stable non-aromatic monocyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond.
  • Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Unless otherwise stated specifically in the specification, a cycloalkyl group may be optionally substituted,
  • Cycloalkylalkyl refers to a radical of the formula - B D where R B is an alkylene chain as defined above and is a cycloalkyl radical as defined above. Unless stated otherwise specifically in the specification, a cycloalkylalkyl group may be optionally substituted.
  • Halo or halogen refers to brorao, chloro, fluoro or iodo.
  • Haloalkyl refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difiuoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1 ,2-difluoroethyl, 3-bromo-2-fiuoropropy[, 1 ,2-dibromoethyi, and the like. Unless stated otherwise specifically in the specification, a haloalkyl group may be optionally substituted.
  • Heterocyclyl or “heterocyclic ring” refers to a stable 3- to 18-membered non-aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heterocyclyl radical is a monocyclic ring system; and the heterocyclyl radical may be partially or fully saturated.
  • heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyi, [l,3]dithianyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpliQllnyl, 2-oxopiperazinyl, 2-oxopiperidinyL 2-oxopyrroiidinyl, oxazolidmyl, piperidinyi, piperazinyi, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1 -oxo-thiomorph.olinyl, and IJ -dioxo-thiomorphoiinyl. Unless stated otherwise specifically in the specification, a heterocyclyl,
  • ⁇ '-heterocyc!y refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. Unless stated otherwise specifically in the specification, an N-heterocyelyl group may be optionally substituted.
  • Heterocyclylalkyl refers to a radical of the formula -R B R E where .
  • R is an alkvlene chain as defined above and R E is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the afkyf radical at the nitrogen atom. Unless stated otherwise specifically in the specification, a heterocyclylalkyl group may be optionally substituted.
  • Heteroaryl refers to a 5- to 14-membered monocyclic ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Examples include, but are not limited to, azepinyl, furanyl, furanonyl, isothiazolyl, imidazolyl, isoxazolyl, oxadiazoiyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1- oxidopyridinyl, 1 -oxidopyrimidinyl, 1-oxidopyrazinyi, 1 -oxidopyridazinyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, thiazolyl, thiadiazolyi, triazolyl, tetrazolyl, triazinyl, thiopheny
  • N-heteroaryl refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl. radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. Unless stated otherwise specifically in the specification, an A/-heteroaryl group may be optionally substituted.
  • Heteroarylalkyl refers to a radical of the formula --R B R F where R B is an alkylene chain as defined above and Rp is a heteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylalkyl group may be optionally substituted.
  • substituted means any of the above groups (i.e., alkyl, alkylene, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloaikylalkyl, haloalkvl, heterocyclyl, N-heterocyciyl, heterocyclylalkyl, heteroaryl, .V-heteroaryl and/or heteroaryialkyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, CI, Br
  • RG and R H are the same or different and independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryi, aralkyl, cycloaikyi, cycloalkylalkyl, haloalkyl, heterocyclyi, N-heterocyclyl, heterocyelylalkyl, heteroaryl, N-heteroaryl and/or heteroaryialkyl.
  • Substituted further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, iraino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloaikyi, cycloalkylalkyl, haloalkyl, heterocyclyi, ⁇ -heterocyclyl, heterocvclyiaikyi, heteroaryl, ⁇ -heteroaryl and/or heteroaryialkyl group, in addition, each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.
  • protecting group refers to a labile chemical moiety which is known in the art to protect reactive groups including without limitation, hydroxyl and amino groups, against undesired reactions during synthetic procedures. Hydroxy! and amino groups protected wit a protecting group are referred to herein as “protected hydroxy! groups” and “protected amino groups”, respectively. Protecting groups are typically used selectively and/or orthogonally to protect sites during reactions at other reactive sites and can then be removed to leave the unprotected group as is or available for further reactions. Protecting groups as known in the art are described generally in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999).
  • groups are protected or present as a precursor that will be inert to reactions that modify other areas of the parent molecule for conversion into their final groups at an appropriate time. Further representative protecting or precursor groups are discussed in Agrawal, et al., Protocols for Oligonucleotide Conjugates, Eds, Humana Press; New Jersey, 1994; Vol. 26 pp. 1-- 72. Examples of "hydroxy!
  • protecting groups include, but are not limited to, t-butyl, t- butoxymethyl, methoxymethyl, tetrahydropyranyl, 1-ethoxyethyl, l-(2- chloroethoxy)ethyl, 2-trimethylsilylethyl, ⁇ -chlorophenyl, 2,4-dinitrophenyl, benzyl, 2,6-dichiorobenzyi, diphenylmethyl, p-nitrobenzyl, triphenylmethyl, trimethyisiiyl, triethylsilyl, t-butyidimethylsiiyi, t-butyldiphenylsilyl (TBDPS), triphenyisiiyl, benzoylformate, acetate, chloroacetate, trichloroacetate, triiluoroacetate, pivaloate, benzoate, p-phenylbenzoate, 9-fiuorenylmethyl carbon
  • amino protecting groups include, but are not limited to, carbamate - protecting groups, such as 2-trirnethylsilylethoxycarbonyi (Teoc), 1 -methyl- 1 -(4- biphenyiyl)ethoxyearhonyl (Bpoc), t-butoxycarbonyl (BOC), allyloxycarbonyl (Alloc), 9-fluorenylmethyloxycarbonyl (Fmoc), and benzyloxycarbonyl (Cbz); amide protecting groups, such as formyi, acetyl, trihaloacetyl, benzoyl, and nitrophenylacetyi; sulfonarai de-protecting groups, such as 2 ⁇ nitrobenzenesulfonyl; and imine and cyclic imide protecting groups, such as phthalimido and dithiasuccinoyl.
  • carbamate - protecting groups such as 2-trirnethylsilylethoxycarbonyi (T
  • the invention disclosed herein is also meant to encompass all pharmaceutically acceptable compounds of Formula (I) being isotopicaliy-labeled by having one or more atoms re laced by an atom having a different atomic mass or mass number.
  • isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, H C, i3 C, i4 C, 13 N, 15 N, !5 0, 17 0, 18 0, 31 P, 32 P, 5 S, l8 F, Jb Cl, i 3 l, and respectively.
  • radiolabeled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action.
  • Certain isotopically-labeled compounds of Formula (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. " ⁇ , and carbon- 14, i.e. i4 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
  • Substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability. For example, in vivo half-life may increase or dosage requirements may be reduced. Thus, heavier isotopes may be preferred in some circumstances.
  • isotopically-labeled compounds of Formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • the invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterifi cation, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising administering a compound of this invention to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
  • an animal such as rat, mouse, guinea pig, monkey, or to human
  • “Stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • “Mammal” includes humans and both domestic animals such as laboratory animals and household pets ⁇ e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.
  • Optional or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • optionally substituted aryl means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • “Pharmaceutically acceptable carrier, diluent or excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
  • “Pharmaceutically acceptable salt” refers to a salt of a compound that is pharmaceutically acceptable and that possesses (or can be converted to a form that possesses) the desired pharmacological activity of the parent compound.
  • “pharmaceutically acceptable salts” of the compounds disclosed herein include salts derived from an appropriate base, such as an alkali metal (for example, sodium), an alkaline earth metal (for example, magnesium), ammonium and X 4 " (wherein X is C J -C4 alkyl).
  • Pharmaceutically acceptable salts of a nitrogen atom or an amino group include for example salts of organic carboxylic acids such as acetic, benzoic, camphorsulfonic, citric, glucoheptonic, gluconic, lactic, fumaric, tartaric, maleic, malonic, malic, mandelic, isethionic, lactobionic, succinic, 2-napththalenesulfomc, oleic, palmitic, propionic, stearic, and trimethyl acetic acids; organic sulfonic acids, such as methanesulfonic, ethanesulfonic, benzenesuifonic and p-toluenesulfonic acids; and inorganic acids, such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric and sulfamic acids.
  • organic carboxylic acids such as acetic, benzoic, camphorsulfonic, citric, glucohe
  • Pharmaceutically acceptable salts of a compound of a hydroxy group include the anion of said compound in combination with a suitable cation such as Na ; and NX 4 f (wherein is independently selected from H or a C 1 --C 4 alkyl group).
  • Pharmaceutically acceptable salts also include salts formed when an acidic proton present, in the parent compound is replaced by either a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aiurmnum ion; or coordinates with an organic base such as diemano famine, triethanolamine, N-methylg!ucamine and the like. Also included in this definition are ammonium and substituted or quaternized ammonium salts.
  • salts of active ingredients of the compounds disclosed herein will typically be pharmaceutically acceptable, i.e. they will be salts derived from a physiologically acceptable acid or base.
  • salts of acids or bases which are not pharmaceutically acceptable may also find use, for example, in the preparation or purification of a compound of Formula (I) or another compound of the invention. All salts, whether or not derived from a physiologically acceptable acid or base, are within the scope of the present invention.
  • Metal salts typically are prepared by reacting the metal hydroxide with a compound of this invention.
  • metal salts which are prepared in this way are salts containing Li + , Na + , and K + .
  • a less soluble metal salt can be precipitated from the solution of a more soluble salt by addition of the suitable metal compound.
  • compositions herein comprise compounds disclosed herein in their un-ionized, as well as zwitterionic form, and combinations with stoichiometric amounts of water as in hydrates.
  • solvate refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent.
  • the solvent may be water, in which case the solvate may be a hydrate.
  • the solvent may be an organic solvent.
  • the compounds of the present invention may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding soivated forms.
  • the compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent.
  • a “pharmaceutical composition” refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans.
  • a medium includes ail pharmaceutically acceptable carriers, diluents or excipients therefor.
  • Effective amount refers to an amount of a compound according to the invention, which when administered to a patient in need thereof, is sufficient to effect treatment for disease-states, conditions, or disorders for which the compounds have utilit . Such an amount would be sufficient to elicit the biological or medical response of a tissue system, or patient that is sought by a researcher or clinician.
  • the amount of a compound according to the invention which constitutes a therapeutically effective amount will vary depending on such factors as the compound and its biological activity, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of the treatment, the type of disease-state or disorder being treated and its severity, dmgs used in combination with or coincidentally with the compounds of the invention, and the age, body weight, general health, sex and diet of the patient.
  • a therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their own knowledge, the state of the art, and this disclosure.
  • treatment is intended to mean the administration of a compound or composition according to the present invention to alleviate or eliminate symptoms of HIV infection and/or to reduce viral load in a patient.
  • treatment also encompasses the administration of a compound or composition according to the present invention post-exposure of the individual to the vims but before the appearance of symptoms of the disease, and/or prior to the detection of the virus in the blood, to prevent the appearance of symptoms of the disease and/or to prevent the virus from reaching detectibie levels in the blood, and the administration of a compound or composition according to the present invention to prevent perinatal transmission of HIV from mother to baby, by administration to the mother before giving birth and to the child within the first days of life.
  • antiviral agent as used herein is intended to mean an agent (compound or biological) that is effective to inhibit the formation and/or replication of a virus in a human being, including but not limited to agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a human being.
  • inhibitor of HIV replication is intended to mean an agent capable of reducing or eliminating the ability of HIV to replicate in a host cell, whether in vitro, ex vivo or in vivo.
  • the compounds of the invention, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (5)- or, as (D)- or (L)- for amino acids.
  • the present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms.
  • Optically active (+) and (-), (2?)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization.
  • stereoisomer refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
  • the present invention contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
  • a “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule.
  • the present invention includes tautomers of any said compounds.
  • a “prodrug” refers to a compound that is chemically designed to efficiently liberate the parent dmg after overcoming biological barriers to oral delivery.
  • the present invention includes prodrugs of the compounds of Formula (I).
  • X is -O- or -NZ 3 - or -CHZ 3 -;
  • W is -CHZ 2 -;
  • Z J , Z 2 and Z 3 are each, independently, hydrogen or C 1-3 alkyl, or wherein Z 1 and Z 2 or Z 1 and Z 3 , taken together, form -L- wherein L is -C(R a ) 2 ⁇ , -C(R a ) 2 C(R a ) 2 -, -C(R a ) 2 C(R a ) 2 C(RV, or -C(R a ) 2 C(R a ) 2 C(R a ) 2 C(R a ) 2 -, wherein at least one of Z 1 and Z 2 or Z 5 and Z ' , taken together, form -L-;
  • Z 4 is a bond, -CH 2 -, or -CH 2 CH 2 -;
  • Y' and Y "6 are each, independently, hydrogen.
  • R 1 is phenyl substituted with one to three halogens;
  • each R a is, independently, hydrogen, halo, hydroxy! or C. ⁇ alkyL
  • compounds having the following f ormula (I l-B):
  • I. is -C(R a ) 2 -.
  • I, is ⁇ C(R a ) 2 C(R a ) 2 C(R a ) 2 -.
  • each R a is hydrogen.
  • one R is methyl and each remaining R. a is hydrogen.
  • one R a is halogen and each remaining R a is hydrogen.
  • two R a are halogen and each remaining R a is hydrogen.
  • one R a is halogen and each remaining R a is hydrogen.
  • X is -0-. In another embodiment, X is -NZ 3 -. In another embodiment, X is -NH-. 16, In another embodiment, X is -CHZ 3 - and Z 1 and Z J , taken together, form -L-. In a further embodiment, 7 is hydrogen. In another embodiment, X is -CH 2 -.
  • Z is a bond or -CH 2 -. In another embodiment,
  • Z 4 is -CH 2 -. In another embodiment, Z 4 is a bond.
  • Y 1 and Y 2 are each independently hydrogen, methyl or trifluoromethyl.
  • R 1 is substituted with one halogen.
  • R 1 is 4-fluorophenyi or 2-fluoropheiiyL
  • R 1 is substituted with two halogens.
  • R 1 is 2,4-d.ifiuorophenyl, 2,3-difIuorophenyl, 2,6-difIuorophenyl, 3- fluoro-4-chlorophenyl, 3,4-difluorophenyl, 2-fj.uoro-4-chloroph.enyl, or 3,5- difluorophenyl.
  • R 1 is 2,4-difiuorophenyl.
  • R ! is substituted with three halogens.
  • R 1 is 2,4,6-trifluorophenyl or 2,3,4-trifluorophenyl. In still a further embodiment, R 1 is 2,4,6-trifluorophenyl.
  • a pharmaceutical composition comprising a compound of any one of the Formulas (I), ( ⁇ - ⁇ ), ( ⁇ - ⁇ ), or (II-C), as noted above, or a stereoisomer or pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier, diluent or excipient.
  • Another embodiment is provided comprising a method of treating an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof.
  • Another embodiment is provided comprising a method of treating or preventing an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof.
  • the use of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, for the treatment of an HIV infection in a human having or at risk of having the infection is provided.
  • the use of a compound of any one of the Formulas (I), (I l-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, for the treatment or prevention of an HIV infection in a human having or at risk of having the infection is provided.
  • the use of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, for use in the therapeutic treatment of an HIV infection is provided.
  • the use of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, for use in the prophylactic or therapeutic treatment of an HIV infection is provided.
  • X is -O- or -NZ 3 - or -CHZ 3 -;
  • W is -()- or -NZ 2 - or -CHZ 2 -;
  • Z 1 , Z 2 and ⁇ ⁇ are each, independently, hydrogen, Ci-salkyl or C 1 -3 haloalkyl, or wherein Z 1 and Z z or Z 1 and Z J , taken together, form -L- wherein L is
  • Z 4 is a bond or -( ⁇ , - . - C I I C f I - , -CH 2 CH 2 CH 2 -, -CH 2 OCH 2 -, -CH 2 NR a CH 2 -, -CH 2 SCH 2 -,-CH 2 S(0)CH 2 - or -CH 2 SG 2 CH 2 -;
  • Y 1 and Y 2 are each, independently, hydrogen or or Y 1 and Y , together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms, wherein the carbocyclic or heterocyclic ring is optionally substituted with one or more R a ;
  • R 3 ⁇ 4 is optionally substituted aryl or optionally substituted heteroaryl; and each R a is, independently, hydrogen, halo, hydroxy! or or wherein two R a groups, together with the carbon atom to which they are attached, form
  • W is -CHZ ' -.
  • Z 1 and Z" or Z 1 and Z 3 taken together, form -L-.
  • compounds are provided having one of the following Formulas ( ⁇ - ⁇ ), (II-B), or (I!-C):
  • L is -C(R a ) 2 C(R a ) 2 -, -C(R a ) 2 C(R a ) 2 C(R a ⁇ 2-
  • Y 1 and Y% together with the carbon atom to which they are attached form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
  • compounds having one of the following Fomiulas (III-A), (III-B), (III-C) or (III-D):
  • Z 1 and Z 3 are each, independently, hydrogen or
  • compounds are provided having one of the following Formulas (III-E), ( ⁇ -F), ( ⁇ -G) or (III ⁇ H):
  • compounds are provided having one of the followmg Fomiulas (IV-AA), (TV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG) or (IV- AH):
  • L is -C(R a )2-, -C(R a ) 2 C(R a ) 2 -, -C(R a ) 2 CiR a ) 2 C(R a ) 2
  • compounds are provided having one of the following Formulas (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG) or (IV-BH):
  • I . is -C(R a ) 2 ⁇ , -C(R a ) 2 C(R a ) 2 -,
  • each R a is hydrogen.
  • one R a is methyl and each remaining R a is hydrogen.
  • one R a is halogen and each remaining R a is hydrogen. In still a further embodiment, two R a are halogen and each remaining R a is hydrogen. In still a further embodiment, one R a is halogen and each remaining R a is hydrogen.
  • L is -C(R a ) 2 OC(R a ) 2 -, -C(R a ) 2 NR a C(R a ) 2 ⁇ , -C(R a ) 2 SC(R a ) 2 - > -C(R a ) 2 S(0)C(R a ) 2 -, or -C(R a ) 2 S0 2 C(R a ) 2 -.
  • L is -C(R a ) 2 OC(R a ) 2 -.
  • each R is hydrogen.
  • one R is methyl and each remaining R a is hydrogen, in still a further embodiment, one R a is halogen and each remaining R a is hydrogen. In still a further embodiment, two R a are halogen and each remaining R a is hydrogen. In still a further embodiment, one R d is halogen and each remaining R is hydrogen .
  • X is -0-. In a further embodiment, Z" is hydrogen. In another embodiment, X is - ⁇ , In another embodiment, is -NH-. In another embodiment, X is -CHZ 3 -. In another embodiment, X is -CH? ⁇ .
  • Z 4 is a bond or ⁇ CH 2 ⁇ . In another embodiment, Z 4 is -CH ? -. In another embodiment, Z 4 is a bond.
  • Y 1 and are each independently hydrogen, methyl or trifluoromethyi.
  • R 1 is substituted with one halogen.
  • R 1 is 4-fiuorophenyl or 2-fluorophenyl.
  • R 1 is phenyl. In another embodiment, R 1 is pyridinyl.
  • R ! is substituted with at least one halogen.
  • R 1 is substituted with one halogen.
  • R 1 is 4-fluorophenyi or 2-fluorophenyl.
  • R 1 is substituted with two halogens.
  • R 1 is 2,4-difluorophenyl, 2,3-difluorophenyl, 2,6-d.ifluorophenyl, 3- fl.uoro-4-chlorophenyl, 3,4-difluorophenyl, 2-fl.uoro-4-chloroph.enyl, or 3,5- difluorophenyl.
  • R 1 is 2,4-difluorophenyl.
  • R ! is substituted with three halogens.
  • R 1 is 2,4,6-trifluorophenyl or 2,3,4-trifluorophenyl. In still a further embodiment, R 1 is 2,4,6-trifluorophenyl.
  • R 1 is 3-trifluoromethyl-4-fluorophenyl or 2- cyclopropoxy-4-fluorophenyl.
  • a pharmaceutical composition comprising a compound of any one of Formulas (I), (II-A.), (II-B), (I !-C), ( ⁇ - ⁇ ), (HI-B), (II l-C), (III-D), (II l-E), (III-F), (III-G), (III-H), (IV-AA), ( I V- ⁇ ). ( IV- ⁇ .
  • Another embodiment comprising a method of treating an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of Formulas (I), (II-A), (II-B), (II-C), (IIl-A), (III-B), (III-C), (III-D), (III-E), (III-F), (III-G), (IIl-H), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof.
  • Another embodiment comprising a method of treating or preventing an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of Formulas (I), (I I -A), (II-B), (II-C), (III-A), (III-B), (III-C), (III-D), (III-E), (III-F), (III-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), ( IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof.
  • IV-AA (IV-AA), (IV- AB), (TV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), ( I V-Al l h (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof, for use in the prophylactic or therapeutic treatment of an HIV infection is provided.
  • the Compound 3 of Example 3 may be shown as:
  • the compounds described herein are administered as a raw chemical or are formulated as pharmaceutical compositions.
  • Pharmaceutical compositions disclosed herein include a compound of Formula (]) and one or more of: a pharmaceutically acceptable carrier, diluent or excipient.
  • the compound of Formula (I) is present in the composition in an amount which is effective to treat a particular disease or condition of interest.
  • the activity of compounds of Formula (I) can be determined by one skilled in the art, for example, as described in the Examples below. Appropriate concentrations and dosages can be readily determined by one skilled in the art.
  • a compound of Formula (I) is present in the pharmaceutical composition in an amount from about 25 mg to about 500 mg.
  • a compound of Formula (I) is present in the pharmaceutical composition in an amount of about 100 mg to about 300 mg. In certain embodiments, a compound of Formula (I) is present in the pharmaceutical composition in an amount of about 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg or about 500 mg.
  • compositions of the invention are prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and in specific embodiments are formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • compositions of the invention are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient.
  • Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aerosol form may hold a plurality of dosage units.
  • composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings described herein.
  • compositions disclosed herein are prepared by methodologies well known in the pharmaceutical art.
  • a pharmaceutical composition intended to be administered by injection is prepared by combining a compound of the invention with sterile, distil led water so as to form a solution
  • a surfactant is added to facilitate the formation of a homogeneous solution or suspension.
  • Surfactants are compounds that non-covalentiy interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.
  • the compounds of the invention are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed ; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy.
  • a method for treating or preventing an HIV infection in a human having or at risk of having the infection comprising administering to the human a therapeutical!)' effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with a therapeutically effective amount of one or more additional therapeutic agents.
  • compositions comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with one or more additional therapeutic agents, and a pharmaceutically acceptable carrier, diluent or excipient are provided.
  • combination pharmaceutical agents comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with one or more additional therapeutic agents are provided.
  • the additional therapeutic agent may be an anti-HIV agent.
  • the additional therapeutic agent is selected from the group consisting of HIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucleotide inhibitors of reverse transcriptase, HIV mtegrase inhibitors, HIV non- catalytic site (or allosteric) integrase inhibitors, entry inhibitors (e.g., CCR5 inhibitors, gp41 inhibitors (i.e., fusion inhibitors) and CD4 attachment inhibitors), CXCR4 inhibitors, gpl20 inhibitors, G6PD and NADH-oxidase inhibitors, compounds that target the HIV capsid ("capsid inhibitors"; e.g., capsid polymerization inhibitors or capsid disrupting compounds such as those disclosed in WO 2013/006738 (Gilead Sciences), US 2013/0
  • HIV protease inhibitors selected from the group consisting of amprenavir, atazanavir, fosamprenavir, indinavir, lopinavir, ritonavir, nelfinavir, saquinavir, tipranavir, brecanavir, darunavir, TMC-126, TMC-1 14, mozenavir (DMP- 450), JE-2147 (AG1776), L-756423, RO0334649, KNI-272, DPC-681, DPC-684, GW640385X, DG17, PPL-10G, DG35, and AG 1859;
  • HIV non-nucleoside or non-nucleotide inhibitors of reverse transcriptase selected from the group consisting of capravirine, emivirine, delaviridine, efavirenz, nevirapine, (+) calanolide A, etravirine, GW5634, DPC-083, DPC-961 , DPC- 963, MIV-150, TMC-120, rilpivirene, BILR 355 BS, VRX 840773, lersivirine (UK- 453061), RDEA806, KM023 and MK-1439;
  • HIV nucleoside inhibitors of reverse transcriptase selected from the group consisting of zidovudine, emtricitabine, didanosine, stavudine, zaicitabine, lamivudine, abacavir, amdoxovir, elvucitabine, alovudine, MIV-210, ⁇ -FTC, D-d4FC, emtricitabine, phosphazide, fozivudine tidoxil, apricitibine (AVX754), KP-1461, GS- 9131 (Giiead Sciences) and fosalvudine tidoxil (formerly HDP 99.0003);
  • HI V nucleotide inhibitors of reverse transcriptase selected from the group consisting of tenofovir, tenofovir disoproxil fumarate, tenofovir alafenarnide fumarate (Giiead Sciences), GS-7340 (Giiead Sciences), GS-9148 (Giiead Sciences), adefovir, adefovir dipivoxii, CMX-001 (Chimerix) or CMX-157 (Chimerix);
  • HIV integrase inhibitors selected from the group consisting of curcumm, derivatives of curcumin, chicoric acid, derivatives of chicoric acid, 3,5- dicaffeoylquitiic acid, derivatives of 3,5-dicaffeoylquinic acid, aurintricarboxylic acid, derivatives of aurintricarboxylic acid, caffeic acid phenethyl ester, derivatives of caffeic acid phenethyl ester, tyrphostin, derivatives of tyrphostin, quercetin, derivatives of quercetin, S-1360, AR-177, L-870812, and L-870810, raltegravir, BMS-538158, GS 364735C, BMS-707035, MK-2048, BA 01 1 , elvitegravir, dolutegravir and GSK- 744;
  • NICKI aliosteric, integrase inhibitors
  • NCINI aliosteric, integrase inhibitors
  • BI-224436 CX0516, CX05045, CX14442
  • compounds disclosed in WO 2009/062285 Boehrmger Ingelheim
  • WO 2010/130034 Boehringer Ingelheim
  • WO 2013/159064 Gilead Sciences
  • WO 2012/145728 Gilead Sciences
  • WO 2012/003497 Gilead Sciences
  • WO 2012/003498 WO 2012/003498
  • gp41 inhibitors selected from the group consisting of enfuvirtide, sifuvirfide, albuvirtide, FB006M, and TRI- 1144;
  • CCR5 inhibitors selected from the group consisting of aplaviroc, vicriviroc, maraviroc, cenicriviroc, PRO- 140, 1 CB15050, PF-232798 (Pfizer), and CCR5mAb004;
  • CD4 attachment inhibitors selected from the group consisting of ibaiizumab (TMB-355) and BMS-068 (BMS-663068);
  • pharmacokinetic enhancers selected from the group consisting of cobicistat and SPI-452;
  • PA-457 (bevirimat), HRG214, VGX-410, KD-247, AMZ 0026, CYT 99007A-221 HIV, DEBIO-025, BAY 50-4798, MDXOIO (ipilimumab), PBS 1 19, ALG 889, and PA-
  • a compound disclosed herein, or a pharmaceutically acceptable salt thereof is combined with two, three, four or more additional therapeutic agents. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with two additional therapeutic agents. In other embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with three additional therapeutic agents. In further embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with four additional therapeutic agents.
  • the two, three four or more additional therapeutic agents can be different therapeutic agents selected from the same class of therapeutic agents, or they can be selected from different classes of therapeutic agents.
  • a compound disclosed herein, or a pharmaceutically acceptable salt thereof is combined with an HIV nucleotide inhibitor of reverse transcriptase and an HIV non-nucleoside inhibitor of reverse transcriptase.
  • a compound disclosed herein, or a pharmaceutically acceptable salt thereof is combined with an HIV nucleotide inhibitor of reverse transcriptase, and an HIV protease inhibiting compound.
  • a compound disclosed herein, or a pharmaceutically acceptable salt thereof is combined with an HIV nucleotide inhibitor of reverse transcriptase, an HIV non-nucleoside inhibitor of reverse transcriptase, and an HIV protease inhibiting compound.
  • a compound disclosed herein, or a pharmaceutically acceptable salt thereof is combined with an HIV nucleotide inhibitor of reverse transcriptase, an HI non-nucleoside inhibitor of reverse transcriptase, and a pharmacokinetic enhancer.
  • the components of the composition are administered as a simultaneous or sequential regimen.
  • the combination may be administered in two or more administrations.
  • a compound disclosed herein is combined with one or more additional therapeutic agents in a unitar dosage form for simultaneous administration to a patient, for example as a solid dosage form for oral administration.
  • a compound disclosed herein is administered with one or more additional therapeutic agents.
  • Co-administration of a compound disclosed herein with one or more additional therapeutic agents generally refers to simultaneous or sequential administration of a compound disclosed herein and one or more additional therapeutic agents, such that therapeutically effective amounts of the compound disclosed herein and one or more additional therapeutic agents are both present in the body of the patient.
  • Co-administration includes administration of unit dosages of the compounds disclosed herein before or after administration of unit dosages of one or more additional therapeutic agents, for example, administration of the compound disclosed herein within seconds, minutes, or hours of the administration of one or more additional therapeutic agents.
  • a unit dose of a compound disclosed herein is administered first, followed within seconds or minutes by administration of a unit dose of one or more additional therapeutic agents.
  • a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of a compound disclosed herein within seconds or minutes.
  • a unit dose of a compound disclosed herein is administered first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of one or more additional therapeutic agents.
  • a unit dose of one or more additional therapeutic agents is administered first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of a compound disclosed herein.
  • Schemes 1-3 are provided as further embodiments of the invention and illustrate general methods which were used to prepare compounds having Formula (I) and which can be used to prepare additional compound having Fonnula (I),
  • Al can be converted to amide A2 with an appropriate amine and a coupling reagent such as HATU or EDCI.
  • A2 can be converted to A3 with a strong acid such as methanesulfonic acid.
  • A3 can be converted to either A5 or A4 by heating with an appropriate cyclic diamine or cyclic aminoalcohol followed by methyl deprotection with a reagent such as magnesium bromide.
  • Al can be converted to A6 by treatment with a strong acid suc as methanesulfonic acid.
  • A6 can be condensed with an appropriate cyclic diamine or cyclic aminoalcohol followed by methyl deprotection with a reagent such as magnesium bromide to form either A7 or A8 respectively.
  • A7 or A8 can be converted into amides A5 and A4 by treatment with an appropriate amine and a coupling reagent such as HATU or EDCI followed by methyl deprotection with a reagent such as magn esi urn bromide .
  • Bl (as described in WO2012/018065) is condensed with diamine under reflux condition to give B2.
  • B2 is hydrolyzed and coupled with an amine by an amide- forming method to afford product B3 upon removal of a benzyl protecting group.
  • WO20.i l 1 19566 is incorporated herein by reference in its entirety.
  • a suspension of Intermediate 5-A (24.8 mg, 0.080 mmol ), l-(2,4- difluoropheny].)cyclopropanamine HC1 salt (5-15, 21.9 mg, 0.107 mmol), and HATU (48 mg, 0.126 mmol) in CH 2 C1 2 (2 mL) was stirred at ambient temperature as N,N- diisopropylethyl amine (DIPEA) (0.1 mL, 0.574 mmol) was added.
  • DIPEA N,N- diisopropylethyl amine
  • reaction mixture was diluted with ethyl acetate before washing with 10% aqueous citric acid solution (xl) and saturated aqueous NaHC0 3 solution (xl). After the aqueous fractions were extracted with ethyl acetate (x l), the organic fractions were combined, dried (MgS0 4 ), and concentrated.
  • Methyl 5-(2,4-difluorobenzy lcarbamoyl)- 1 -(2,2-dihydroxyethy i)-3- metboxy-4-oxo-l,4-dibydropyridine-2-carboxylate (l-C, 0,050 g, 0.121 mmol), (lS,3R)-3-aminocyclohexanol (0,028 g, 0.243 mmol) and potassium carbonate (0.034 g, 0.243 mmol) were suspended in acetonitrile (0.95 mL) and heated to 90 °C for 0.5 hour.
  • reaction mixture was cooled and partitioned between dichloromethane and HQ (0.2 M aq). The layers were separated and the aqueous layer was extracted again with dichloromethane. The combined organic layers were treated with a small amount of acetonitrile, dried over sodium sulfate, filtered and concentrated.
  • Methyl 5-(2,4-difluorobenzylcarbamoyl)- 1 -(2,2-dihydroxyethyl)-3- metiioxy-4-oxo-l ,4-diiiydropyridine-2-carboxyiate (l-C, 97.5 mg, 0.236 mmol) was treated with acetoiiitrile (1.9 mL), acetic acid (0, 1 mL), potassium carbonate (145 mg, 1 .05 fflfflol), and (8) ⁇ piperidm-3-amme dihydrochloride (82 rag, 0.472 rnmol). The reaction mixture was sealed and heated to 90 °C.
  • the aqueous phase was thrice extracted into dichloromethane and the combined organic phases were combmed, dried over MgS0 4 , filtered, concentrated.
  • the crude product was dissolved into acetonitrile (2 mL) and magnesium bromide (96.5 mg, 0.52 mmol) was added. The mixture was reseated and heated to 50 °C. After 80 minutes, the reaction mixture was quenched with ⁇ 5 mL of 0.2M HC1 (aq), the pH adjusted to - 10, diluted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile : water, 0.1% TFA) afforded Compound 20. !
  • the reaction mixture was sealed and heated to 90 °C. After 60 minutes, the reaction mixture was cooled partitioned between brine and dichloromethane. The aqueous phase was thrice extracted into dichloromethane and the combined organic phases were combined, dried over MgS0 4 , filtered, and concentrated. The crude product was dissolved into acetonitrile (2 mL) and magnesium bromide (74 mg, 0.4 mrnol) was added. The mixture was resealed and heated to 50 °C. After 100 minutes, the reaction mixture was quenched with 0.2 ⁇ 1 HQ (aq), diluted with brine, and thrice extracted into DCM.
  • the aqueous phase was thrice extracted into dichioromethane and the combined organic phases were combined, dried over MgS0 4 , filtered, and concentrated.
  • the crude product was dissolved into acetoniirile (2 mL) and magnesium bromide (91 mg, 0.49 mmol ) was added. The mixture was resealed and heated to 50 °C. After 100 minutes, the reaction mixture was quenched with 0.2M HCl(aq), diluted with brine, and thrice extracted into DCM. HPLC purification (acetoniirile: water, 0.1% TFA) afforded Compound 22.
  • Compound 29-B (13 mg, 14%) was prepared from compound 27-B (87 mg, 0,187 mmol) and the aminoalcohol 29-A (45 mg, 0.391 mmol) in a manner similar to that described in step 1 of the synthesis of compound 28-B.
  • Compound 31-B (123 mg, 81%) was prepared from compound 27-B (150 mg, 0.322 mmol) and the aminoalcohol 31-A (70,3 mg, 0.695 mmol) in a manner similar to that described in step 1 and 2 of the synthesis of compound 30 ⁇ B.
  • Cuprous cyanide (290 mg, 3,27 mmol) was suspended in 3.3 mL THF and cooled to -78 °C.
  • a 1.6M solution of Me Li (4.1 mL, 6.56 mmol) in diethyl ether was added dropwise, the reaction solution was allowed to warm to room temperature over the course of 2 hours, and recooled to -78 °C.
  • Intermediate 39-A (190 mg) was separated by chiral Prep-HP LC on a Lux Cellulose-2 column using 9: 1 ACN:MeOH as eluent to afford Intermediates 39-B (first eluting peak) and 40-A (second during peak) in enantioenriched form.
  • Intermediate 39-B (absolute stereochemistry confinned by XRay crystallography), Chiral HPLC retention time ::: 3.98 minutes (Lux Cellulose-2 IC, 150 x 4.6 mm, 2 mL/min 9: 1 ACN:MeOH).
  • Step 5 a
  • the reaction mixture was allowed to warm to room temperature, 2,4-difluorobenzonitrile (3.48 g, 25 mmol) was added portionwise, and reaction temperature raised to 95 °C.
  • the reaction solution was cooled to room temperature after stirring for 18 hours, diluted with ethyl acetate, washed twice with water and twice with brine, dried over MgS0 4 , filtered, and concentrated onto silica gel.
  • Example 61 was prepared analogously to Example 60, substituting (1 S,3S,4R) ⁇ tert-b tyl 3-(aminomethy].)-2-azabicyclo[2.2. l]heptane-2-carboxylate (prepared in Example 55) for 41-E, and (3-ch].oro-2,4-dif3.uorophenyl)methanamine for (2,3- dichlorophenyl)methanamine.
  • 'H-NMR 400 MHz, DMSO-d6) ⁇ 1 1.85 (s, 1 H), 10.45 (t, IH), 8.40 (s, I H), 7.37 (td, I I I ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Molecular Biology (AREA)
  • AIDS & HIV (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Compounds for use in the treatment of human immunodeficiency virus (HIV) infection are disclosed. The compounds have the following Formula (I): including stereoisomers and pharmaceutically acceptable salts thereof, wherein R1, X, W, Y1, Y2, Ζ1 and Z4 are as defined herein. Methods associated with preparation and use of such compounds, as well as pharmaceutical compositions comprising such compounds, are also disclosed.

Description

POLYCYCLIC-CARBAMOYLPYRIDONE COMPOUNDS AND THEIR
PHARMACEUTICAL USE
CROSS-REFERENCE TO RELATED APPLICATIONS
This patent application claims the benefit under 35 U.8.C. § 119(e) of U.S. Provisional Patent Application No. 61/745,375, filed December 21, 2012, U.S. Provisional Patent Application No. 61/788,397, filed March 15, 2013, and U.S. Provisional Patent Application No. 61/845,803, filed July 12, 2013. The foregoing applications are incorporated herein by reference in their entireties. BACKGROUND
Field
Compounds, compositions, and methods for the treatment of human immunodeficiency vims (HIV) infection are disclosed. In particular, novel poly cyclic carbamoylpyridone compounds and methods for their preparation and use as therapeutic or prophylactic agents are disclosed.
Description of the Related Art
Human immunodeficiency virus infection and related diseases are a major public health problem worldwide. Human immunodeficiency vims type 1 (HTV-1) encodes three enzymes which are required for viral replication: reverse transcriptase, protease, and integrase. Although drugs targeting reverse transcriptase and protease are in wide use and have shown effectiveness, particularly when employed in combination, toxicity and development of resistant strains have limited their usefulness (Palella, et al. N. Engl. J Med, (1998) 338:853 -860; Richman, D. D. Nature (2001) 410:995-1001).
Pregnane X receptor (PXR) is a nuclear receptor that is one of the key regulators of enzymes involved in metabolism and elimination of small molecules from the body. Activation of PXR is known to up-regulate or induce the production of metabolic enzymes such as cytochrome P450 3A4 (CYP3A4) as well as enzymes involved in transport, such as OATP2 in the liver and intestine (Endocrine Reviews (2002) 23(5):ό87-702). When one drug causes the up-regulation of these and other enzymes by activation of PXR, this can reduce the absorption and/or exposure of a coadministered drug that is susceptible to the up-regulated enzymes. To minimize the risk of this type of drug-drug interaction, it is desirable to minimize PXR activation. Further, it is known that PXR is activated by many different classes of molecules [Endocrine Reviews (2002) 23(5):687-702). Thus for drugs that will be co-administered with other drugs, it is important to test for and minimize PXR activation.
Transporters have been identified as playing a role in the pharmacokinetic, safety and efficacy profile or drugs, and certain drug-drug interactions are mediated by transporters. See, Giacomini KM, et al. ""Membrane transporters in drug development," NaLRev Drug Discov. 9: 215-236, 2010; Zhang L, et al. "Transporter- Mediated Drug-Drug Interactions," Clin. Pharm. Ther. 89(4):481-484 (2011). -One tranporter, the organic cation transporter 2 (OCT2; SLC22A2), is a member of the solute carrier (SLC) super-family of transporters and is primarily localized on the basolateral membrane of the renal proximal tubule, OCT2, in concert with apical expressed multidrug and toxin extrusion (MATE) transporters 1 and 2-K, is believed to form the major cationic secretion pathway in the kidney and has been shown to transport endogenous compounds including creatinine and xenobiotics including metformin. Inhibition of OCT2 can thus lead to increased levels of serum creatinine and the potential for increased levels of other OCT2 substrates, it is important as well to test and reduce OCT2 inhibition of drugs.
A goal of antiretroviral therapy is to achieve viral suppression in the HIV infected patient. Treatment guidelines published by the United States Department of Health and Human Services provide that achievement of viral suppression requires the use of combination therapies, i.e., several drugs from at least two or more dr g classes. (Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV- 1 -infected adults and adolescents. Department of Health and Human Sendees. Available at littp://aidsinfo.nih.gov/ConteiitFiles/AduItandAdolescentGL.pdf. Section accessed March 14, 2013.) In addition, decisions regarding the treatment of HIV infected patients are complicated when the patient requires treatment for other medical conditions (Id. at E-.12). Because the standard of care requires the use of multiple different drugs to suppress HIV, as well as to treat other conditions the patient may be experiencing, the potential for drug interaction is a criterion for selection of a drug regimen. As such, there is a need for antiretroviral therapies having a decreased potential for drug interactions.
Accordingly, there is a need for new agents that inhibit the replication of HIV and that minimize PXR activation when co-administered with other drugs.
BRIEF SUMMARY
The present invention is directed to novel polycyclic carbamoylpyridone compounds, having antiviral activity, including stereoisomers and pharmaceutical [y acceptable salts thereof, and the use of such compounds in the treatment of HIV infections. The compounds of the invention may be used to mhibit the activity of HIV integrase and may be used to reduce HIV replication.
In one embodiment of the present invention, compounds having the following Formula (I) are provided:
Figure imgf000004_0001
G) or a stereoisomer or pharmaceutically acceptable salt thereof,
wherein:
X is ·()·· or -NZ3- or -CHZ3-;
W is -CHZ2-;
Z1, Zz and Z3 are each, independently, hydrogen or Cj-3alkyl, or wherein Z1 and Z2 or Z1 and Z3, taken together, form -L- wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, or -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, wherein at least one of Z1 and Z2 or Zx and Z3, taken together, form -L-; Z is a bond, -CH2-, or ·( !! ( I !>·;
Y1 and Y2 are each, independently, hydrogen, Cj-aalkyl or
Figure imgf000005_0001
R¾ is phenyl substituted with one to three halogens; and
each R3 is, independently, hydrogen, halo, hydroxyl or ('· salky].
In another embodiment of the present invention, compounds having the following Fomiula (I) are provided:
Figure imgf000005_0002
(I) or a stereoisomer or pharmaceutically acceptable salt thereof,
wherein:
X is -O- or -NZ3- or -CHZ3-;
W is -O- or -NZ2- or -CHZ2-:
Z1, Z2 and Z3 are each, independently, hydrogen or C1-3alkyl, or wherein Z1 and Z2 or Zl and Z3, taken together, form -L- wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2OC(Ra)2-,
-C(Ra)2NRaC(Ra)2~, ~C(Ra)2SC(Ra)2-, ~C(Ra)2S(0)C(Ra)2~, ~C(Ra)2S02C(Ra)2~, -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2OC(Ra)2-, -C(R3)2NRaC(R3)2C(Ra)2-,
-C(Ra)2C(Ra)2NR3C(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C{R! ('{I<J)>SC{RJ)>-.
{( :')>S(0 (Κ'')-(·(Μ:'}'··. ({κ·!}>(·(Κ , (())(·(κ·!}>·, - (\ } O (R:,} (\ }^ -C(Ra)2C(Ra)2S02C(Ra)2-, -C(Ra)2802NRaC(Ra)2- or -C(Ra)2NRaS02C(Ra)2-;
Z is a bond or -CH2-, ·('!!.■( H.-. -CH2CH2CH2-, -CH2OCH2-, -CH2NRaCH2-, -CH2SCH2-,-CH2S(0)CH2- or -CH2S02CH2-;
Y1 and Yi are each, independently, hydrogen,
Figure imgf000005_0003
or Ci.3h.al0al.kyl, or Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms, wherein the carbocyciic or heterocyclic ring is optionally substituted with one or more R ;
R5 is optionally substituted aryl or optional!)' substituted heteroaryl; and each Ra is, independently, hydrogen, halo, hydroxyl or C1.4alkyl, or wherein two Ra groups, together with the carbon atom to which they are attached, form
=0, and
wherein at least one of: (i) Z1 and Z2 or Z1 and ZJ, taken together, form -L-; or (ii) Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyciic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
In another embodiment, a pharmaceutical composition is provided comprising a compound having Formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
The invention also provides the use of a pharmaceutical composition as described hereinabove for the treatment of an HIV infection in a human being having or at risk of having the infection.
In another embodiment, a method of using a compound having Formula (I) in therapy is provided. In particular, a method of treating the proliferation of the HIV virus, treating AIDS, or delaying the onset of AIDS or ARC symptoms in a mammal (e.g., a human) is provided, comprising administering to the mammal a compound having Formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
In another embodiment, use of a compound of Formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for the treatment of an HIV infection in a human being having or at risk of having the infection is disclosed.
In another embodiment, the use of a compound of Formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of an HIV infection in a human being having or at risk of having the infection is disclosed.
In another embodiment, an article of manufacture comprising a composition effective to treat an HIV infection; and packaging material comprising a label which indicates that the composition can be used to treat infection by HIV is disclosed. Exemplary compositions comprise a compound of Formula (I) according to this invention or a pharmaceutically acceptable salt thereof.
In still another embodiment, a method of inhibiting the replication of HIV is disclosed. The method comprises exposing the virus to an effective amount of the compound of Formula (I), or a salt thereof, under conditions where replication of HIV is inhibited.
In another embodiment, the use of a compound of Formula (I) to inhibit the activity of the HIV integrase enzyme is disclosed.
In another embodiment, the use of a compound of Formula (I), or a salt thereof, to inhibit the replication of HIV is disclosed.
Other embodiments, objects, features and advantages will be set forth in the detailed description of the embodiments that follows, and in part will be apparent from the description, or may be learned by practice, of the claimed invention. These objects and advantages will be realized and attained by the processes and compositions particularly pointed out in the written description and claims hereof. The foregoing Summary has been made with the understanding that it is to be considered as a brief and general synopsis of some of the embodiments disclosed herein, is provided solely for the benefit and convenience of the reader, and is not intended to limit in any manner the scope, or range of equivalents, to which the appended claims are lawfully entitled.
DETAI LED DESCRIPTION
In the following description, certain specific details are set forth in order to provide a thorough understanding of various embodiments of the invention. However, one skilled in the art will understand that the invention may be practiced without these details. The description below of several embodiments is made with the understanding that the present disclosure is to be considered as an exemplification of the claimed subject matter, and is not intended to limit the appended claims to the specific embodiments illustrated. The headings used throughout this disclosure are provided for convenience only and are not to be construed to limit the claims in any way. Embodiments illustrated under any heading may be combined with embodiments illustrated under any other heading. Definitions
Unless the context requires otherwise, throughout the present specification and claims, the word "comprise" and variations thereof, such as, "comprises" and "comprising" are to be construed in an open, inclusive sense, that is as "including, but not limited to".
Reference throughout this specification to "one embodiment" or "an embodiment" means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present invention. Thus, the appearances of the phrases "in one embodiment" or "in an embodiment" in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments.
Unless the context requires otherwise, reference throughout this specification to "a compound of Formula (I)" or "compounds of Formula (I)" refers to ail embodiments of Formula (I), including, for example, compounds of Formulas (II-A), (II-B), (II-C), (III-A), (ΠΙ-Β), (III-C), (III-D), (II -E), (III-F), (ITI-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (TV-AD), (IV-AE), (IV-AF), (TV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as well as the specific compounds disclosed herein.
"Amino" refers to the -N¾ radical.
"Cyano" refers to the -CN radical.
"Hydroxy" or "hydroxy!" refers to the -OH radical.
"Iraino" refers to the :::NH substituent.
"Nitro" refers to the -N02 radical.
"Oxo" refers to the =0 substituent.
"Thioxo" refers to the =;:S substituent.
"Alkyl" refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated {i.e., contains one or more double and/or triple bonds), having from one to twelve carbon atoms (Ci-Cj? alkyl), preferably one to eight carbon atoms (Cj-Cg alkyl) or one to six carbon atoms (Ci-Ce alkyl), and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, H-propyl, l-rnethylethyl (zso-propyl), «-butyl, n-pentyl, 1 ,1-dirnethylethyl (/-butyl), 3-methylhexyl, 2-meth.ylh.exyi, etherryl, prop-l-enyl, but-l -enyl, pent-l-enyl, penta-l.,4-dienyl, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted.
"Alkylene" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, n-butylene, ethenylene, propenylene, n-butenylene, propynylene, «~butynylene, and the like. The alkylene chain is attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkylene chain may be optionally substituted.
"Alkoxy" refers to a radical of the formula -ORA where RA is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxy group may be optionally substituted.
"Alkylamino" refers to a radical of the formula -NHRA or -NRARA where each is, independently, an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylamino group may be optionally substituted.
Thioalkyl" refers to a radical of the formula -SRA where A is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a thioalkyl group may be optionally substituted.
"Aryl" refers to a monocylic hydrocarbon ring system radical comprising hydrogen and 6 to 18 carbon atoms. Aryl radicals include, but are not limited to, aryl radicals derived from benzene. Unless stated otherwise specifically in the specification, the term "aryl" or the prefix "ar-" (such as in "aralkyl") is meant to include aryl radicals that are optionally substituted. "Araikyi" refers to a radical of the formula -RB-RC where RB is an alkylene chain as defined above and c is one or more aryl radicals as defined above, for example, benzyl. Unless stated otherwise specifically in the specification, an aralkyl group may be optionally substituted.
"Cycloalkyl" or "carbocyclic ring" refers to a stable non-aromatic monocyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond. Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Unless otherwise stated specifically in the specification, a cycloalkyl group may be optionally substituted,
"Cycloalkylalkyl" refers to a radical of the formula - B D where RB is an alkylene chain as defined above and is a cycloalkyl radical as defined above. Unless stated otherwise specifically in the specification, a cycloalkylalkyl group may be optionally substituted.
"Halo" or "halogen" refers to brorao, chloro, fluoro or iodo. "Haloalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difiuoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1 ,2-difluoroethyl, 3-bromo-2-fiuoropropy[, 1 ,2-dibromoethyi, and the like. Unless stated otherwise specifically in the specification, a haloalkyl group may be optionally substituted.
"Heterocyclyl" or "heterocyclic ring" refers to a stable 3- to 18-membered non-aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. In the embodiments disclosed herein, the heterocyclyl radical is a monocyclic ring system; and the heterocyclyl radical may be partially or fully saturated. Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyi, [l,3]dithianyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpliQllnyl, 2-oxopiperazinyl, 2-oxopiperidinyL 2-oxopyrroiidinyl, oxazolidmyl, piperidinyi, piperazinyi, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1 -oxo-thiomorph.olinyl, and IJ -dioxo-thiomorphoiinyl. Unless stated otherwise specifically in the specification, a heterocyclyl group may be optionally substituted.
"Λ'-heterocyc!y!" refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. Unless stated otherwise specifically in the specification, an N-heterocyelyl group may be optionally substituted.
"Heterocyclylalkyl" refers to a radical of the formula -RBRE where .R is an alkvlene chain as defined above and RE is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the afkyf radical at the nitrogen atom. Unless stated otherwise specifically in the specification, a heterocyclylalkyl group may be optionally substituted.
"Heteroaryl" refers to a 5- to 14-membered monocyclic ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Examples include, but are not limited to, azepinyl, furanyl, furanonyl, isothiazolyl, imidazolyl, isoxazolyl, oxadiazoiyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1- oxidopyridinyl, 1 -oxidopyrimidinyl, 1-oxidopyrazinyi, 1 -oxidopyridazinyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, thiazolyl, thiadiazolyi, triazolyl, tetrazolyl, triazinyl, thiophenyl, and thienyl. Unless stated otherwise specifically in the specification, a heteroaryl group may be optionally substituted.
"N-heteroaryl" refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl. radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. Unless stated otherwise specifically in the specification, an A/-heteroaryl group may be optionally substituted.
"Heteroarylalkyl" refers to a radical of the formula --RBRF where RB is an alkylene chain as defined above and Rp is a heteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylalkyl group may be optionally substituted.The term "substituted" used herein means any of the above groups (i.e., alkyl, alkylene, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloaikylalkyl, haloalkvl, heterocyclyl, N-heterocyciyl, heterocyclylalkyl, heteroaryl, .V-heteroaryl and/or heteroaryialkyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, CI, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylaniines, alkylarylamines, diai lamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyi groups, dialkylarylsilyi groups, alkyldiarylsilyl groups, and triaryl.sil.yl groups; and other heteroatoms in various other groups, "Substituted" also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbony!, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. For example, "substituted" includes any of the above groups in which one or more hydrogen atoms are replaced with -NRQRH,
Figure imgf000012_0001
-N RT ;C( (»ORn. -\R<X ( \ RTT)M ( IR„. -NRGSG2RH, ··()( '( 0)\Κ(π. -OR, ,- -SRG, -SORG, -S02RG, -OS02RG, -S02ORG, =NS02Rg, and -S02NRGRH. "Substituted also means any of the above groups in which one or more hydrogen atoms are replaced with -C(=0)RG, -Π ()} }R(.. -C(==0)NRGRH, -C:H2S02RG, -CH2S02NRGRH. in the foregoing, RG and RH are the same or different and independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryi, aralkyl, cycloaikyi, cycloalkylalkyl, haloalkyl, heterocyclyi, N-heterocyclyl, heterocyelylalkyl, heteroaryl, N-heteroaryl and/or heteroaryialkyl. "Substituted" further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, iraino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloaikyi, cycloalkylalkyl, haloalkyl, heterocyclyi, Λ-heterocyclyl, heterocvclyiaikyi, heteroaryl, Λ-heteroaryl and/or heteroaryialkyl group, in addition, each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.
The term "protecting group," as used herein, refers to a labile chemical moiety which is known in the art to protect reactive groups including without limitation, hydroxyl and amino groups, against undesired reactions during synthetic procedures. Hydroxy! and amino groups protected wit a protecting group are referred to herein as "protected hydroxy! groups" and "protected amino groups", respectively. Protecting groups are typically used selectively and/or orthogonally to protect sites during reactions at other reactive sites and can then be removed to leave the unprotected group as is or available for further reactions. Protecting groups as known in the art are described generally in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999). Generally, groups are protected or present as a precursor that will be inert to reactions that modify other areas of the parent molecule for conversion into their final groups at an appropriate time. Further representative protecting or precursor groups are discussed in Agrawal, et al., Protocols for Oligonucleotide Conjugates, Eds, Humana Press; New Jersey, 1994; Vol. 26 pp. 1-- 72. Examples of "hydroxy! protecting groups" include, but are not limited to, t-butyl, t- butoxymethyl, methoxymethyl, tetrahydropyranyl, 1-ethoxyethyl, l-(2- chloroethoxy)ethyl, 2-trimethylsilylethyl, ρ-chlorophenyl, 2,4-dinitrophenyl, benzyl, 2,6-dichiorobenzyi, diphenylmethyl, p-nitrobenzyl, triphenylmethyl, trimethyisiiyl, triethylsilyl, t-butyidimethylsiiyi, t-butyldiphenylsilyl (TBDPS), triphenyisiiyl, benzoylformate, acetate, chloroacetate, trichloroacetate, triiluoroacetate, pivaloate, benzoate, p-phenylbenzoate, 9-fiuorenylmethyl carbonate, mesylate and tosylate. Examples of "amino protecting groups" include, but are not limited to, carbamate - protecting groups, such as 2-trirnethylsilylethoxycarbonyi (Teoc), 1 -methyl- 1 -(4- biphenyiyl)ethoxyearhonyl (Bpoc), t-butoxycarbonyl (BOC), allyloxycarbonyl (Alloc), 9-fluorenylmethyloxycarbonyl (Fmoc), and benzyloxycarbonyl (Cbz); amide protecting groups, such as formyi, acetyl, trihaloacetyl, benzoyl, and nitrophenylacetyi; sulfonarai de-protecting groups, such as 2~nitrobenzenesulfonyl; and imine and cyclic imide protecting groups, such as phthalimido and dithiasuccinoyl.
The invention disclosed herein is also meant to encompass all pharmaceutically acceptable compounds of Formula (I) being isotopicaliy-labeled by having one or more atoms re laced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H, 3H, HC, i3C, i4C, 13N, 15N, !50, 170, 180, 31P, 32P, 5S, l8F, JbCl, i 3l, and respectively. These radiolabeled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action. Certain isotopically-labeled compounds of Formula (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. "Ή, and carbon- 14, i.e. i4C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability. For example, in vivo half-life may increase or dosage requirements may be reduced. Thus, heavier isotopes may be preferred in some circumstances.
Substitution with positron emitting isotopes, such as ' . 18F, l 50 and !&N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy, isotopically-labeled compounds of Formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
The invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterifi cation, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising administering a compound of this invention to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
"Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. "Mammal" includes humans and both domestic animals such as laboratory animals and household pets {e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.
"Optional" or "optionally" means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
"Pharmaceutically acceptable carrier, diluent or excipient" includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
"Pharmaceutically acceptable salt" refers to a salt of a compound that is pharmaceutically acceptable and that possesses (or can be converted to a form that possesses) the desired pharmacological activity of the parent compound. Examples of "pharmaceutically acceptable salts" of the compounds disclosed herein include salts derived from an appropriate base, such as an alkali metal (for example, sodium), an alkaline earth metal (for example, magnesium), ammonium and X4 " (wherein X is CJ-C4 alkyl). Pharmaceutically acceptable salts of a nitrogen atom or an amino group include for example salts of organic carboxylic acids such as acetic, benzoic, camphorsulfonic, citric, glucoheptonic, gluconic, lactic, fumaric, tartaric, maleic, malonic, malic, mandelic, isethionic, lactobionic, succinic, 2-napththalenesulfomc, oleic, palmitic, propionic, stearic, and trimethyl acetic acids; organic sulfonic acids, such as methanesulfonic, ethanesulfonic, benzenesuifonic and p-toluenesulfonic acids; and inorganic acids, such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric and sulfamic acids. Pharmaceutically acceptable salts of a compound of a hydroxy group include the anion of said compound in combination with a suitable cation such as Na ; and NX4 f (wherein is independently selected from H or a C1--C4 alkyl group). Pharmaceutically acceptable salts also include salts formed when an acidic proton present, in the parent compound is replaced by either a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aiurmnum ion; or coordinates with an organic base such as diemano famine, triethanolamine, N-methylg!ucamine and the like. Also included in this definition are ammonium and substituted or quaternized ammonium salts. Representative non-limiting lists of pharmaceutically acceptable salts can be found in S.M. Berge et ai, J. Pharoia Sci., 66(1), 1-19 (1977), and Remington: The Science and Practice of Pharmacy, R. Hendrickson, ed,, 21st edition, Lippincott, Williams & Wilkins, Philadelphia, PA, (2005), at p. 732, Table 38-5, both of which are hereby incorporated by reference herein.
For therapeutic use, salts of active ingredients of the compounds disclosed herein will typically be pharmaceutically acceptable, i.e. they will be salts derived from a physiologically acceptable acid or base. However, salts of acids or bases which are not pharmaceutically acceptable may also find use, for example, in the preparation or purification of a compound of Formula (I) or another compound of the invention. All salts, whether or not derived from a physiologically acceptable acid or base, are within the scope of the present invention.
Metal salts typically are prepared by reacting the metal hydroxide with a compound of this invention. Examples of metal salts which are prepared in this way are salts containing Li+, Na+, and K+. A less soluble metal salt can be precipitated from the solution of a more soluble salt by addition of the suitable metal compound.
In addition, salts may be formed from acid addition of certain organic and inorganic acids, e.g., HQ, HBr, H2SO . H3PO4 or organic sulfonic acids, to basic centers, typically amines. Finally, it is to be understood that the compositions herein comprise compounds disclosed herein in their un-ionized, as well as zwitterionic form, and combinations with stoichiometric amounts of water as in hydrates.
Often crystallizations produce a solvate of the compound of the invention. As used herein, the term "solvate" refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent. The solvent may be water, in which case the solvate may be a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding soivated forms. The compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent.
A "pharmaceutical composition" refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes ail pharmaceutically acceptable carriers, diluents or excipients therefor.
"Effective amount" or "therapeutically effective amount" refers to an amount of a compound according to the invention, which when administered to a patient in need thereof, is sufficient to effect treatment for disease-states, conditions, or disorders for which the compounds have utilit . Such an amount would be sufficient to elicit the biological or medical response of a tissue system, or patient that is sought by a researcher or clinician. The amount of a compound according to the invention which constitutes a therapeutically effective amount will vary depending on such factors as the compound and its biological activity, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of the treatment, the type of disease-state or disorder being treated and its severity, dmgs used in combination with or coincidentally with the compounds of the invention, and the age, body weight, general health, sex and diet of the patient. Such a therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their own knowledge, the state of the art, and this disclosure.
The term "treatment" as used herein is intended to mean the administration of a compound or composition according to the present invention to alleviate or eliminate symptoms of HIV infection and/or to reduce viral load in a patient. The term "treatment" also encompasses the administration of a compound or composition according to the present invention post-exposure of the individual to the vims but before the appearance of symptoms of the disease, and/or prior to the detection of the virus in the blood, to prevent the appearance of symptoms of the disease and/or to prevent the virus from reaching detectibie levels in the blood, and the administration of a compound or composition according to the present invention to prevent perinatal transmission of HIV from mother to baby, by administration to the mother before giving birth and to the child within the first days of life. The term "antiviral agent" as used herein is intended to mean an agent (compound or biological) that is effective to inhibit the formation and/or replication of a virus in a human being, including but not limited to agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a human being.
The term "inhibitor of HIV replication" as used herein is intended to mean an agent capable of reducing or eliminating the ability of HIV to replicate in a host cell, whether in vitro, ex vivo or in vivo.
The compounds of the invention, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (5)- or, as (D)- or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (2?)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centres of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.
A "stereoisomer" refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present invention contemplates various stereoisomers and mixtures thereof and includes "enantiomers", which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
A "tautomer" refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present invention includes tautomers of any said compounds. A "prodrug" refers to a compound that is chemically designed to efficiently liberate the parent dmg after overcoming biological barriers to oral delivery. In certain embodiments, the present invention includes prodrugs of the compounds of Formula (I). Compounds
As noted above, in one embodiment of the present invention, compounds having antiviral activity are provided, the compounds having the following Formula (I):
Figure imgf000019_0001
(0 or a stereoisomer or pharmaceutically acceptable salt thereof,
wherein:
X is -O- or -NZ3- or -CHZ3-;
W is -CHZ2-;
ZJ , Z2 and Z3 are each, independently, hydrogen or C1-3alkyl, or wherein Z1 and Z2 or Z1 and Z3, taken together, form -L- wherein L is -C(Ra)2~, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(RV, or -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, wherein at least one of Z1 and Z2 or Z5 and Z' , taken together, form -L-;
Z4 is a bond, -CH2-, or -CH2CH2-;
Y' and Y"6 are each, independently, hydrogen. Chalk ! or d-sha oalkyl; R1 is phenyl substituted with one to three halogens; and
each Ra is, independently, hydrogen, halo, hydroxy! or C.^alkyL
In another embodiment, compounds are provided having the following Formula (II -A):
Figure imgf000020_0001
(II-A)
In another embodiment, compounds are provided having the following formula (I l-B):
Figure imgf000020_0002
(II-B
In another embodiment, compounds are provided having the following
Formula (II-C):
Figure imgf000020_0003
(II-C)
In another embodiment, I. is -C(Ra)2-. In a further embodiment, L C(Ra)2C(Ra)2~. In still a further embodiment, I, is ~C(Ra)2C(Ra)2C(Ra)2-. In still further embodiment, each Ra is hydrogen. In still a further embodiment, one R is methyl and each remaining R.a is hydrogen. In still a further embodiment, one Ra is halogen and each remaining Ra is hydrogen. In still a further embodiment, two Ra are halogen and each remaining Ra is hydrogen. In still a further embodiment, one Ra is halogen and each remaining Ra is hydrogen.
In another embodiment, X is -0-. In another embodiment, X is -NZ3-. In another embodiment, X is -NH-. 16, In another embodiment, X is -CHZ3- and Z1 and ZJ, taken together, form -L-. In a further embodiment, 7 is hydrogen. In another embodiment, X is -CH2-.
In another embodiment, Z is a bond or -CH2-. In another embodiment,
Z4 is -CH2-. In another embodiment, Z4 is a bond.
In another embodiment, Y1 and Y2 are each independently hydrogen, methyl or trifluoromethyl.
In another embodiment, R1 is substituted with one halogen. In a further embodiment, R1 is 4-fluorophenyi or 2-fluoropheiiyL
In another embodiment, R1 is substituted with two halogens. In a further embodiment, R1 is 2,4-d.ifiuorophenyl, 2,3-difIuorophenyl, 2,6-difIuorophenyl, 3- fluoro-4-chlorophenyl, 3,4-difluorophenyl, 2-fj.uoro-4-chloroph.enyl, or 3,5- difluorophenyl. In still a further embodiment, R1 is 2,4-difiuorophenyl.
In another embodiment, R! is substituted with three halogens. In a further embodiment, R1 is 2,4,6-trifluorophenyl or 2,3,4-trifluorophenyl. In still a further embodiment, R1 is 2,4,6-trifluorophenyl.
In one embodiment, a pharmaceutical composition is provided comprising a compound of any one of the Formulas (I), (Π-Α), (Π-Β), or (II-C), as noted above, or a stereoisomer or pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier, diluent or excipient.
Another embodiment, is provided comprising a method of treating an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof. Another embodiment is provided comprising a method of treating or preventing an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof.
In another embodiment, the use of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, for the treatment of an HIV infection in a human having or at risk of having the infection is provided. In another embodiment, the use of a compound of any one of the Formulas (I), (I l-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, for the treatment or prevention of an HIV infection in a human having or at risk of having the infection is provided.
In another embodiment, the use in medical therapy of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, is provided.
In another embodiment, the use of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, for use in the therapeutic treatment of an HIV infection is provided. In another embodiment, the use of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, for use in the prophylactic or therapeutic treatment of an HIV infection is provided.
As further noted above, in another embodiment of the present invention, compounds having antiviral activity are provided, the compounds having the following Formula (I):
Figure imgf000022_0001
ω or a stereoisomer or pharmaceutically acceptable salt thereof,
wherein:
X is -O- or -NZ3- or -CHZ3-;
W is -()- or -NZ2- or -CHZ2-;
Z1, Z2 and ΖΛ are each, independently, hydrogen, Ci-salkyl or C1 -3haloalkyl, or wherein Z1 and Zz or Z1 and ZJ, taken together, form -L- wherein L is
•C{ a )..··. -C(Ra)2C(Ra)2-, ·<·( ΚΊ.-( ( ! ") :( '{ Ra )..··. -C(Ra)2C(Ra)2C(Ra)2C(R
-C(Ra)2OC(Ra)2-» ~C(Ra)2NRaC(Ra)2~, -C(Ra)2SC(Ra)2-, -C(Ra)2S(0)C(Ra)2-, ~C(Ra)2S02C(Ra)2~, -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2QC(Ra)2-, -C(Ra)2NR3C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2~, -C(Ra)2S(0)C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2S(0)C(Ra)2-, -C ^SO .H R'W K' -C(Ra)2C( )2S02C(Ra)2-, -C(Ra)2S02NRaC(Ra)2- or -C(Ra)2NRaS02C(Ra)2 ;
Z4 is a bond or -( Ή, - . - C I I C f I - , -CH2CH2CH2-, -CH2OCH2-, -CH2NRaCH2-, -CH2SCH2-,-CH2S(0)CH2- or -CH2SG2CH2-;
Y1 and Y2 are each, independently, hydrogen or
Figure imgf000023_0001
or Y1 and Y , together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms, wherein the carbocyclic or heterocyclic ring is optionally substituted with one or more Ra;
R¾ is optionally substituted aryl or optionally substituted heteroaryl; and each Ra is, independently, hydrogen, halo, hydroxy! or
Figure imgf000023_0002
or wherein two Ra groups, together with the carbon atom to which they are attached, form
=Ό, and
wherein at least one of: (i) Z5 and 7 or Z! and Z' taken together, form -L-; or
(ii) Y 5 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
In another embodiment, W is -CHZ'-.
In another embodiment, Z1 and Z" or Z1 and Z3, taken together, form -L-.
In another embodiment, compounds are provided having one of the following Formulas (Π-Α), (II-B), or (I!-C):
Figure imgf000024_0001
(II-A)
Figure imgf000024_0002
; or
(II-B)
Figure imgf000024_0003
il!-O wherein L is -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra}2-
-C(Ra)2C(Ra)2C(Ra)2C(Ra) -C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-
-C(Ra)2S(0)C(Ra}2-, -C(Ra)2S02C(Ra)2-, ··('{ Ra )■()(( R''} ('(R:' }.-
-( (R").( Κ ·),()( YR,! 5-, -C(Ra)2 RaC(Ra)2C(Ra): -C(Ra)2C(Ra)2NRaC(Ra)2-
-C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2S(0)C(Ra)2C(Ra)2-
-C(Ra)2C(Ra)2S(0)C(Ra)2- -C(Ra)2S02C(Ra)2C(Ra> -C(Ra)2C(Ra)2S02C(Ra)2-
-C(Ra)2S02NRaC(Ra)2- or -C(Ra)2NRaS02C(Ra)2- In another embodiment, Y1 and Y% together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
In another embodiment, compounds are provided having one of the following Fomiulas (III-A), (III-B), (III-C) or (III-D):
Figure imgf000025_0001
(III-C)
Figure imgf000026_0001
< 1 I !--! ) wherein Z1 and Z3 are each, independently, hydrogen or
Figure imgf000026_0002
In another embodiment, compounds are provided having one of the following Formulas (III-E), (Πί-F), (ΠΙ-G) or (III~H):
Figure imgf000026_0003
(III-F)
Figure imgf000027_0001
(III-G)
Figure imgf000027_0002
(III-H) wherein Z and Z" are each, independently, hydrogen or Cj-aalkyl.
In another embodiment, both (i) Z1 and ZS or Z.} and Z3, taken together, form -L-, and (if) Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
In another embodiment, compounds are provided having one of the followmg Fomiulas (IV-AA), (TV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG) or (IV- AH):
Figure imgf000027_0003
(IV-AA)
Figure imgf000028_0001
Figure imgf000028_0002
Figure imgf000028_0003
Figure imgf000028_0004
(IV-AE)
Z /
Figure imgf000029_0001
(IV-AF)
Figure imgf000029_0002
wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2CiRa)2C(Ra)2
-C(Ra)2C(Ra}2C(Ra)2C(Ra)2- -C(Ra)2OC(Ra)2-, ~C(Ra)2NRaC(Ra}2-, -C(Ra)2SC(Ra)2
-C(Ra)2S(0)C(Ra)2-, -C(Ra)2S02C(Ra)2-, -C(Ra)2OC(Ra)2C(Ra)2
-C(Ra)2(:;(Ra)2oc(Ri,)2-, -QRa)2NRaC(Rn)2C(Ra)2-s -C(Ra)2C( R3)2NRaC(Ra')2
-C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2~, ~C(Ra)2S(0)C(Ra)2C(Ra)2 -C(Ra)2C(Rn)2S(0)C(Ra)2-, -C(Ra)2S02C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2S02C(Ra)2
-C(Ra)2S02NRaC(Ra)2- or -C(Ra)2NRaS02C(Ra)2 In anoiher embodiment, compounds are provided having one of the following Formulas (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG) or (IV-BH):
Figure imgf000030_0001
(IV-BC)
Figure imgf000031_0001
Figure imgf000032_0001
(IV-BH) wherein L is -C(Ra)2~, -C{R! ('(RJ) -. ~C(Ra)2C(Ra)2C(R3)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, ··( ( R") -()( ( R-:),-. -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(0)C(Ra)2-, -C(Ra)2S02C(Ra)2-, -C(Ra)2OC(Ra)2C(Ra)2-,
-C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, ~C(Ra)2C(Ra)2NRaC(Ra)2~, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(0)C(Ra)2C(Ra)2-,
-C(Ra)2C(Ra)2S(0)C(Ra)2-, -C( a)2S02C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2S02C(Ra)2-, -C(Ra)2S02NRaC(Ra)2- or -C(Ra)2NRaS02C(Ra)2-.
In another embodiment, I . is -C(Ra)2~, -C(Ra)2C(Ra)2-,
-C(Ra)2C(Ra)2C(Ra)2-, or -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, In a further embodiment, I, is -C(R )2-. In still a farther embodiment, L is -C(R )2C(Ra)?-. In still a further embodiment, L is ~C(R )2C(Ra)2C(Ra)2-. In still a further embodiment, each Ra is hydrogen. In still a further embodiment, one Ra is methyl and each remaining Ra is hydrogen. In still a further embodiment, one Ra is halogen and each remaining Ra is hydrogen. In still a further embodiment, two Ra are halogen and each remaining Ra is hydrogen. In still a further embodiment, one Ra is halogen and each remaining Ra is hydrogen.
In another embodiment, L is -C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2~, -C(Ra)2SC(Ra)2-> -C(Ra)2S(0)C(Ra)2-, or -C(Ra)2S02C(Ra)2-. In a further embodiment, L is -C(Ra)2OC(Ra)2-. In still a further embodiment, each R is hydrogen. In still a further embodiment, one R is methyl and each remaining Ra is hydrogen, in still a further embodiment, one Ra is halogen and each remaining Ra is hydrogen. In still a further embodiment, two Ra are halogen and each remaining Ra is hydrogen. In still a further embodiment, one Rd is halogen and each remaining R is hydrogen . In another embodiment, X is -0-. In a further embodiment, Z" is hydrogen. In another embodiment, X is -ΝΖ , In another embodiment, is -NH-. In another embodiment, X is -CHZ3-. In another embodiment, X is -CH?~.
In another embodiment, Z4 is a bond or ~CH2~. In another embodiment, Z4 is -CH?-. In another embodiment, Z4 is a bond.
In another embodiment, Y1 and are each independently hydrogen, methyl or trifluoromethyi.
In another embodiment, R1 is substituted with one halogen. In a further embodiment, R1 is 4-fiuorophenyl or 2-fluorophenyl.
In another embodiment, R1 is phenyl. In another embodiment, R1 is pyridinyl.
In another embodiment, R! is substituted with at least one halogen.
In another embodiment, R1 is substituted with one halogen. In a further embodiment, R1 is 4-fluorophenyi or 2-fluorophenyl.
In another embodiment, R1 is substituted with two halogens. In a further embodiment, R1 is 2,4-difluorophenyl, 2,3-difluorophenyl, 2,6-d.ifluorophenyl, 3- fl.uoro-4-chlorophenyl, 3,4-difluorophenyl, 2-fl.uoro-4-chloroph.enyl, or 3,5- difluorophenyl. In stil l a further embodiment, R1 is 2,4-difluorophenyl.
In another embodiment, R ! is substituted with three halogens. In a further embodiment, R1 is 2,4,6-trifluorophenyl or 2,3,4-trifluorophenyl. In still a further embodiment, R1 is 2,4,6-trifluorophenyl.
In another embodiment, R1 is 3-trifluoromethyl-4-fluorophenyl or 2- cyclopropoxy-4-fluorophenyl.
In one embodiment, a pharmaceutical composition is provided comprising a compound of any one of Formulas (I), (II-A.), (II-B), (I !-C), (ΠΙ-Α), (HI-B), (II l-C), (III-D), (II l-E), (III-F), (III-G), (III-H), (IV-AA), ( I V-ΛΒ ). ( IV-ΛΟ. (IV-AD), (IV-AE), (IV-AF), (ΪΥ-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (TV-BE), (TV-BF), (IV-BG), and (IV-BH), as noted above, or a stereoisomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
Another embodiment is provided comprising a method of treating an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of Formulas (I), (II-A), (II-B), (II-C), (IIl-A), (III-B), (III-C), (III-D), (III-E), (III-F), (III-G), (IIl-H), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof. Another embodiment is provided comprising a method of treating or preventing an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of Formulas (I), (I I -A), (II-B), (II-C), (III-A), (III-B), (III-C), (III-D), (III-E), (III-F), (III-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), ( IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof.
In another embodiment, the use of a compound of any one of Formulas (I), (II-A), (II-B), (II-C), (III-A), (III-B), (III-C), (III-D), (III-E), (III-F), (III-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof for the treatment of an HIV infection in a human having or at risk of having the infection. In another embodiment, the use of a compound of any one of Formulas (I), (II-A), (Ιί-Β), (II-C), (III-A), (III-B), (I H-C), (III-D), (III-E), (III-F), (III-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (IV -AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof for the treatment or prevention of an HIV infection in a human having or at risk of having the infection.
In another embodiment, the use in medical therapy of a compound of any one of the Formulas (I), (II-A), (II-B), (II-C), (III-A), (III-B), (III-C), (Ili-D), (III-E), (III-F), (III-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof, is provided.
In another embodiment, the use of a compound of any one of the Formulas (I), (II-A), (II-B), (II-C), (III-A), (III-B), (III-C), (III-D), (III-E), (III-F), (ΊΠ-G h (ΙΠ-Η), (IV-AA), f V-AI5). ( I V-ΛΟ. (TV-AD), (I -AE), (IV-AF), (TV-AG), (TV-AH), (TV-BA), (IV-BB), (IV-BC), ( IV-BDh (IV-BE), (IV-BF), (IV-BG ). and
(IV-BH), as noted above, or a pharmaceutical composition thereof, for use in the therapeutic treatment of an HIV infection is provided. In another embodiment, the use of a compound of any one of the Formulas (]), (II-A), (II-B), (II~C), (III-A), (III-B), ( 11 !-C). (ΠΙ-D), (III-E), (III-F), ( l ll -G ). ( 111-! ! ). (IV-AA), (IV- AB), (TV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), ( I V-Al l h (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof, for use in the prophylactic or therapeutic treatment of an HIV infection is provided.
It is understood that any embodiment of the compounds of Formulas (I), (II-A), (II-B), (II-C), (I l l-A ). (III-B), ( I l l-C). (III-D), ( Il l-H i. (III-F), (III-G), (Ili-H), (IV-AA), (TV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (TV-AG), (TV-AH), (TV-BA), (IV-BB), (TV-BC), (IV-BD), (IV-BE), (IV -BF), (IV-BG), and (IV-BH), as set forth above, and any specific substituent set forth herein for a R1, R\ X, W, Y1, Y2, L, Z1, Z% Z3, or Z4 group in the compounds of Formulas (I), (II-A), (II-B), (II-C), (III-A), (III-B), ( Il l-C ). (III-D), (III-E), ί ί ί Ι-Γ ). (III-G), ( I II-I ! ), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (TV-AF), (IV-AG), (TV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as set forth above, may be independently combined with other embodiments and/or substituents of compounds of Formulas (I), (II-A), (II-B), (II-C), (III-A), (III-B), (III-C), (III-D), (III-E), (III-F), (III-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (TV-AF), (TV-AG), (IV-AH), (IV-BA), (TV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), to form embodiments of the inventions not specifically set forth above. In addition, in the event that a list of substitutents is listed for any particular R1, R , X, W, Y1, Y% L, Z1, Z2, Z3, or Z4 in a particular embodiment and/or claim, it is understood that each individual substituent may be deleted from the particular embedment and/or claim and that the remaining list of substituents will be considered to be within the scope of the invention.
As one of skill in the art will appreciate, compounds of Formulas (I), (II-A), (II-B), (II-C), (IV-AA), (IV-AB), (TV-AC), (IV-AD), (IV-AE), (TV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (TV-BH), wherein Z1 and 7 or Z* and Z3, taken together, form -L- may be shown in several different ways. For example, the Compound 3 of Example 3 may be shown as:
Figure imgf000036_0001
Pharmaceutical Compositions
For the purposes of administration, in certain embodiments, the compounds described herein are administered as a raw chemical or are formulated as pharmaceutical compositions. Pharmaceutical compositions disclosed herein include a compound of Formula (]) and one or more of: a pharmaceutically acceptable carrier, diluent or excipient. The compound of Formula (I) is present in the composition in an amount which is effective to treat a particular disease or condition of interest. The activity of compounds of Formula (I) can be determined by one skilled in the art, for example, as described in the Examples below. Appropriate concentrations and dosages can be readily determined by one skilled in the art. In certain embodiments, a compound of Formula (I) is present in the pharmaceutical composition in an amount from about 25 mg to about 500 mg. In certain embodiments, a compound of Formula (I) is present in the pharmaceutical composition in an amount of about 100 mg to about 300 mg. In certain embodiments, a compound of Formula (I) is present in the pharmaceutical composition in an amount of about 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg or about 500 mg.
Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, is carried out via any of the accepted modes of administration of agents for serving similar utilities. The pharmaceutical compositions of the invention are prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and in specific embodiments are formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols. Exemplary routes of administering such pharmaceutical compositions include, without limitation, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal, and intranasal. Pharmaceutical compositions of the invention are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient. Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aerosol form may hold a plurality of dosage units. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 20th Edition (Philadelphia College of Pharmacy and Science, 2000). The composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings described herein.
The pharmaceutical compositions disclosed herein are prepared by methodologies well known in the pharmaceutical art. For example, in certain embodiments, a pharmaceutical composition intended to be administered by injection is prepared by combining a compound of the invention with sterile, distil led water so as to form a solution, in some embodiments, a surfactant is added to facilitate the formation of a homogeneous solution or suspension. Surfactants are compounds that non-covalentiy interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.
The compounds of the invention, or their pharmaceutically acceptable salts, are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed ; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy.
Combination Therapy
In one embodiment, a method for treating or preventing an HIV infection in a human having or at risk of having the infection is provided, comprising administering to the human a therapeutical!)' effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with a therapeutically effective amount of one or more additional therapeutic agents.
In one embodiment, pharmaceutical compositions comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with one or more additional therapeutic agents, and a pharmaceutically acceptable carrier, diluent or excipient are provided.
In one embodiment, combination pharmaceutical agents comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with one or more additional therapeutic agents are provided.
In the above embodiments, the additional therapeutic agent may be an anti-HIV agent. For example, in some embodiments, the additional therapeutic agent is selected from the group consisting of HIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucleotide inhibitors of reverse transcriptase, HIV mtegrase inhibitors, HIV non- catalytic site (or allosteric) integrase inhibitors, entry inhibitors (e.g., CCR5 inhibitors, gp41 inhibitors (i.e., fusion inhibitors) and CD4 attachment inhibitors), CXCR4 inhibitors, gpl20 inhibitors, G6PD and NADH-oxidase inhibitors, compounds that target the HIV capsid ("capsid inhibitors"; e.g., capsid polymerization inhibitors or capsid disrupting compounds such as those disclosed in WO 2013/006738 (Gilead Sciences), US 2013/0165489 (University of Pennsylvania), and WO 2013/006792 (Pharma Resources), pharmacokinetic enhancers, and other drugs for treating HIV, and combinations thereof. In further embodiments, the additional therapeutic agent is selected from one or more of:
( 1 ) HIV protease inhibitors selected from the group consisting of amprenavir, atazanavir, fosamprenavir, indinavir, lopinavir, ritonavir, nelfinavir, saquinavir, tipranavir, brecanavir, darunavir, TMC-126, TMC-1 14, mozenavir (DMP- 450), JE-2147 (AG1776), L-756423, RO0334649, KNI-272, DPC-681, DPC-684, GW640385X, DG17, PPL-10G, DG35, and AG 1859;
(2) HIV non-nucleoside or non-nucleotide inhibitors of reverse transcriptase selected from the group consisting of capravirine, emivirine, delaviridine, efavirenz, nevirapine, (+) calanolide A, etravirine, GW5634, DPC-083, DPC-961 , DPC- 963, MIV-150, TMC-120, rilpivirene, BILR 355 BS, VRX 840773, lersivirine (UK- 453061), RDEA806, KM023 and MK-1439;
(3) HIV nucleoside inhibitors of reverse transcriptase selected from the group consisting of zidovudine, emtricitabine, didanosine, stavudine, zaicitabine, lamivudine, abacavir, amdoxovir, elvucitabine, alovudine, MIV-210, ±-FTC, D-d4FC, emtricitabine, phosphazide, fozivudine tidoxil, apricitibine (AVX754), KP-1461, GS- 9131 (Giiead Sciences) and fosalvudine tidoxil (formerly HDP 99.0003);
(4) HI V nucleotide inhibitors of reverse transcriptase selected from the group consisting of tenofovir, tenofovir disoproxil fumarate, tenofovir alafenarnide fumarate (Giiead Sciences), GS-7340 (Giiead Sciences), GS-9148 (Giiead Sciences), adefovir, adefovir dipivoxii, CMX-001 (Chimerix) or CMX-157 (Chimerix);
(5) HIV integrase inhibitors selected from the group consisting of curcumm, derivatives of curcumin, chicoric acid, derivatives of chicoric acid, 3,5- dicaffeoylquitiic acid, derivatives of 3,5-dicaffeoylquinic acid, aurintricarboxylic acid, derivatives of aurintricarboxylic acid, caffeic acid phenethyl ester, derivatives of caffeic acid phenethyl ester, tyrphostin, derivatives of tyrphostin, quercetin, derivatives of quercetin, S-1360, AR-177, L-870812, and L-870810, raltegravir, BMS-538158, GS 364735C, BMS-707035, MK-2048, BA 01 1 , elvitegravir, dolutegravir and GSK- 744;
(6) HIV non-catalytic site, or aliosteric, integrase inhibitors (NCINI) including, but not limited to, BI-224436, CX0516, CX05045, CX14442, compounds disclosed in WO 2009/062285 (Boehrmger Ingelheim), WO 2010/130034 (Boehringer Ingelheim), WO 2013/159064 (Gilead Sciences), WO 2012/145728 (Gilead Sciences), WO 2012/003497 (Gilead Sciences), WO 2012/003498 (Gilead Sciences) each of which is incorporated by references in its entirety herein;
(7) gp41 inhibitors selected from the group consisting of enfuvirtide, sifuvirfide, albuvirtide, FB006M, and TRI- 1144;
(8) the CXCR4 inhibitor AMD-070;
(9) the entry inhibitor SP01A;
(10) the gpl20 inhibitor BMS-488043;
(1 1) the G6PD and NADH-oxidase inhibitor immunitm;
(12) CCR5 inhibitors selected from the group consisting of aplaviroc, vicriviroc, maraviroc, cenicriviroc, PRO- 140, 1 CB15050, PF-232798 (Pfizer), and CCR5mAb004;
(13) CD4 attachment inhibitors selected from the group consisting of ibaiizumab (TMB-355) and BMS-068 (BMS-663068);
(14) pharmacokinetic enhancers selected from the group consisting of cobicistat and SPI-452; and
(15) other drugs for treating HIV selected from the group consisting of
BAS-100, SPI-452, REP 9, SP-01 A, TNX-355, DES6, ODN-93, ODN-1 12, VGV- I .
PA-457 (bevirimat), HRG214, VGX-410, KD-247, AMZ 0026, CYT 99007A-221 HIV, DEBIO-025, BAY 50-4798, MDXOIO (ipilimumab), PBS 1 19, ALG 889, and PA-
1050040 (PA-040),
and combinations thereof
In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with two, three, four or more additional therapeutic agents. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with two additional therapeutic agents. In other embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with three additional therapeutic agents. In further embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with four additional therapeutic agents. The two, three four or more additional therapeutic agents can be different therapeutic agents selected from the same class of therapeutic agents, or they can be selected from different classes of therapeutic agents. In a specific embodiment, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with an HIV nucleotide inhibitor of reverse transcriptase and an HIV non-nucleoside inhibitor of reverse transcriptase. In another specific embodiment, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with an HIV nucleotide inhibitor of reverse transcriptase, and an HIV protease inhibiting compound. In a further embodiment, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with an HIV nucleotide inhibitor of reverse transcriptase, an HIV non-nucleoside inhibitor of reverse transcriptase, and an HIV protease inhibiting compound. In an additional embodiment, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with an HIV nucleotide inhibitor of reverse transcriptase, an HI non-nucleoside inhibitor of reverse transcriptase, and a pharmacokinetic enhancer.
In certain embodiments, when a compound disclosed herein is combined with one or more additional therapeutic agents as described above, the components of the composition are administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations.
In certain embdoiments, a compound disclosed herein is combined with one or more additional therapeutic agents in a unitar dosage form for simultaneous administration to a patient, for example as a solid dosage form for oral administration.
In certain embodiments, a compound disclosed herein is administered with one or more additional therapeutic agents. Co-administration of a compound disclosed herein with one or more additional therapeutic agents generally refers to simultaneous or sequential administration of a compound disclosed herein and one or more additional therapeutic agents, such that therapeutically effective amounts of the compound disclosed herein and one or more additional therapeutic agents are both present in the body of the patient.
Co-administration includes administration of unit dosages of the compounds disclosed herein before or after administration of unit dosages of one or more additional therapeutic agents, for example, administration of the compound disclosed herein within seconds, minutes, or hours of the administration of one or more additional therapeutic agents. For example, in some embodiments, a unit dose of a compound disclosed herein is administered first, followed within seconds or minutes by administration of a unit dose of one or more additional therapeutic agents. Alternatively, in other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of a compound disclosed herein within seconds or minutes. In some embodiments, a unit dose of a compound disclosed herein is administered first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of one or more additional therapeutic agents. In other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of a compound disclosed herein.
The following Examples illustrate various methods of making compounds of this invention, i.e., compound of Formula (I):
Figure imgf000042_0001
CD wherein R1, X, W, Y5, Y , Z1, 7. or Z" are as defined above, it is understood that one skilled in the art may be able to make these compounds by similar methods or by combining other methods known to one skilled in the art. It is also understood that one skilled in the art would be able to make, in a similar manner as described below, other compounds of Formula (I) not specifically illustrated below by using the appropriate starting components and modifying the parameters of the synthesis as needed. In general, starting components may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and Fluorochem USA, etc. or synthesized according to sources known to those skilled in the art (see, for example, Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition (Wiley, December 2000)) or prepared as described herein.
The following examples are provided for purposes of illustration, not limitation.
EXAMPLES
GENERAL SYNTHETIC SCHEMES
Schemes 1-3 are provided as further embodiments of the invention and illustrate general methods which were used to prepare compounds having Formula (I) and which can be used to prepare additional compound having Fonnula (I),
Figure imgf000043_0001
AS
Figure imgf000043_0002
Figure imgf000043_0003
Figure imgf000043_0004
Al can be converted to amide A2 with an appropriate amine and a coupling reagent such as HATU or EDCI. A2 can be converted to A3 with a strong acid such as methanesulfonic acid. A3 can be converted to either A5 or A4 by heating with an appropriate cyclic diamine or cyclic aminoalcohol followed by methyl deprotection with a reagent such as magnesium bromide.
Alternatively, Al can be converted to A6 by treatment with a strong acid suc as methanesulfonic acid. A6 can be condensed with an appropriate cyclic diamine or cyclic aminoalcohol followed by methyl deprotection with a reagent such as magnesium bromide to form either A7 or A8 respectively. A7 or A8 can be converted into amides A5 and A4 by treatment with an appropriate amine and a coupling reagent such as HATU or EDCI followed by methyl deprotection with a reagent such as magn esi urn bromide .
Figure imgf000044_0001
Bl (as described in WO2012/018065) is condensed with diamine under reflux condition to give B2. B2 is hydrolyzed and coupled with an amine by an amide- forming method to afford product B3 upon removal of a benzyl protecting group.
REPRESENTATIVE COMPOUNDS
Example i
Preparation of Compound 1
N-(2,4-difluorobenzyi)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13a-octahydro-2,5- methanopyrido[ 1 \2':4,5]pyrazmo[2, 1 -b] [ 1 ,3]oxazepine-l 0-carboxamide
Figure imgf000044_0002
ί (+/-)
Figure imgf000045_0001
1 -A 1 -C
Figure imgf000045_0002
1 (+/-)
Step 1
l -(2,2-dimethoxyethyi)-5-methoxy-6-(methoxycarbonyl)-4-oxo-l,4- dihydropyridme-3-carboxylie acid (1-A, 0.300 g, 0.95 mmol), prepared as described in WO201 1/1 19566 A l , was evaporated once from dry toluene, suspended in acetonitri!e (4 mL) and treated with Ν,Ν-diisopropylethylamine (DIPEA) (0.329 mL, 1.90 mmol), 2,4-difluorobenzylamine (0.125 mL, 1 .05 mmol) and HATU (0.433 g, 1.14 mmol). The reaction mixture was stirred for 10 minutes and concentrated. The residue was purified by flash chromatography on silica gel (10 to 60% ethyl acetate :dichloromethane) to afford the compound methyl 5-(2,4-difIuorobenzylearbamoyi)~ 1 -(2,2-d.imeth.oxyeth l)- 3-methoxy-4-oxo- 1 ,4-dihydropyridine-2-carboxylate, 1-B. Ή-NMR (400 MHz, DMSO-d6i δ 10.28 (t, ./ 6.0 Hz, 1 H), 8.46 (s, 1 1 1 ). 7.42 (dd, 15.4, 8.6 Hz, 1 H), 7.24 (m, 1 H), 7.06 (m, 1 H), 4.52 (m, 3H), 4.22 (d, ./ 4.4 Hz, 2H), 3.92 is. M l ). 3.80 (s, 3H),3.29 (d, 6H). LCMS-ESI+ ( n/z): \ M · 1 1 1 calculated for ( ·..·! f.- -J ; · \ () -: 441.15; found: 441.2.
Ste 2
Methyl 5~(2,4~difluorobenzylcarbamoyl)-l-(2,2-dirnethoxyethyl)-3~ methoxy-4-oxo-l. ,4-dihydropyridine-2-carboxylate (1-B, 0.106 g, 0.24 mmol) in acetonitrile (0.9 mL) and acetic acid (0, 1 mL) was treated with methanesulfonic acid (0.005 mL, 0.072 mmol), sealed with a yel low cap, and heated to 70 °C. After 16 hours, the mixture was cooled to afford a crude solution of methyl 5-(2,4- difluorobenzylcarbamoyl)-l-(2,2-dihydroxyethyl)-3-methoxy-4-oxo-l ,4- dihydropyridine-2-carboxyiate, 1-C. LCMS-ESF (m/z): [M+H] calculated for
Figure imgf000046_0001
413.12; found: 413.1.
Steps 3 and 4
Methyl 5-(2,4-difluorobenzylcarbamoyl)- 1 -(2,2-dihydroxyethyl)-3- methoxy-4-oxo-l ,4-dihydropyridine-2-carboxylate (1-C, 0.65 mL of the crude mixture from the previous step, 0.17 mmoi) was treated with acetonitrile (0.65 mL) and cis-3- aminocyclpentanol (0.06 ml..). The reaction mixture was sealed and heated to 90 °C. After 30 minutes, the reaction mixture was cooled and magnesium bromide (0.063 g, 0.34 mmol) was added. The mixture was reseated and heated to 50 °C. After 10 minutes, the reaction mixture was partitioned between dichioromethane and hydrochloric acid (0.2 M aq). The organic layer was removed and the aqueous layer extracted again with dichlormethane. The combined organic layers were dried over sodium sulfate, filtered and concentrated. Prep-HPLC purification (30-70% acetonitrile I water, 0.1% TFA) afforded Compound 1 as a raeemic mixture. 1H-NMR (400 MHz, DMSO-d6) δ 12.45 (br s, I I I ). 10.35 (t, J = 5.8 Hz, i l l ), 8.45 (s, H i ), 7.37 (dd, J 15.4, 8.6 Hz, I H), 7.23 (dt, J = 2.5, 9.9 Hz, 1H), 7.05 (dt, J = 2.2, 8.7 Hz, 1H), 5.43 (dd, 9.6, 4.0 Hz, 1 H), 5.09 (br s, 1H), 4.68 (dd, J = 13.2, 4.0 Hz, 1 H), 4.59 (br s, H I }. 4.53 (m, 21 1 K 4.02 (dd. ./ 12.6, 9.4 Hz), 1.93 (br s, 4H), 1.83 (d. ./ 12.0 Hz), 1.57 (dt, J = 12.2, 3.2 Hz). LCMS-ESf (m/z): [M+H]+ calculated for C^ i HzoFzNsOs : 432.14; found: 432.2.
Examples 2 asid 3
Preparation of Compounds 2 and 3
(2R,5S, 13aR)-N-(2,4-difluorobenzyl)-8-hydroxy-7,9-dioxo
octahydro-2,5-methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b][ 1 ,3]oxazepine- 10-carboxamide (2) and (2S,5R,13aS)-N 2,4-difluorobenzyi)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,l^ octahydro-2,5-methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b][ 1 ,3]oxazepine- 10-carboxamide
(3)
Figure imgf000047_0001
Compound I (16 mg) was separated by chiral HPLC using Chiralpak A.S-H with 1 00% ethanol as eluent to afford Compounds 2 and 3 in enantiomerically enriched form. For Compound 2: LCMS-ESI* (m/z): [M+ll]T~ calculated for C21H2oF2N305: 432.14; found: 432.2, Chiral HPLC retention time = 4.50 minutes (Chiralpak AS-H, 150 x 4.6 mm, 1 mL/min EtOH). For Compound 3: LCMS-ES (m/z) [M+H calculated for C21H20F2N3O5 : 432.14; found: 432.2, Chiral HPLC retention time = 6.84 minutes (Chiralpak AS-H, 150 x 4.6 mm, 1 mL/min EtOH). H- NMR (400 M Hz, DMSQ~d6) δ 12.45 (br s, 1H), 10.35 (t, J ------ 5.8 Hz, 1 H), 8.44 (s, 1 H),
7.37 (dd, J = 15.2, 8.4 Hz, I I I . 7.23 (m, I H), 7.05 (dt, ./ = 1.8 Hz, 8.7 Hz, I H), 5.44 (dd, 9.6, 4.0 Hz), 5.09 (br s, I H), 4.68 (dd, ./ 12.8, 4.0 Hz, I H), 4.59 (br s, I H), 4.53 (m, 2H), 4.02 (dd, J = 12.6, 9.4 Hz, I H), 1.93 (br s, 4H), 1.83 (d, 12.4 Hz, I H), 1.57 (m, IH).
Alternatively, Compound 3 was prepared as follows:
Figure imgf000048_0001
Figure imgf000048_0002
Methyl 5-(2,4-difluorobenzylcarbamoyl)- 1 -(2,2-dihydroxyethyl)-3- methoxy-4-oxo-l ,4-dihydropyridine-2-carboxylate (1-C, 1.2 mmol in 5 mL of 9: 1 acetomtrile: acetic acid containing 0.026 mL methanesulfonic acid) was treated with acetonitrile (5.0 mL) and c'«'-3-ammocyclpentanol (0.24 g, 2.4 mmol). The reaction mixture was sealed and heated to 90 °C. After 30 minutes, the reaction mixture was cooled, treated with potassium carbonate (0.332 g, 2.4 mmol), sealed and reheated to 90 °C. After 15 minutes, the mixture was cooled and partitioned between dichlormethane and hydrochloric acid (0.2 M aqueous). The organic layer was removed and the aqueous solution was extracted again with dichloromethane. The combined organic layers were dried over sodium sulfate (anhydrous), filtered and concentrated. The residue was purified by flash chromatography (0-8% ethanol (containing 1 1% saturated aqueous ammonium hydroxide) in dichloromethane) to afford Intermediate 1-D. LCMS-ES (m/z : i VI · 1 1 1 calculated for C . f i · !· ,X .< 446.15; found: 446.2 Intermediate 1-D (270 rag) was separated by chiral SFC on a 50 mm Chiralpak AD-H column using 50% (1 : 1 rnethanohacetonitrile) in supercritical carbon dioxide as e!uent to afford Intermediates 3-A (first el ting peak) and 3-B (second e luting peak) in enantioenriched form. For 3-A: LCMS-ES (m/z): [M+H]+ calculated for C22H77F2N3O5 : 446.15; found: 446.2. For 3-B: LCMS-ESF (m/z): | VI H i calculated for C22H22F2N3O5: 446.15; found: 446.2.
Intermediate 3-A (0.1 10 g, 0.247 mmol) in acetonitrile (5 mL) was treated portion wise with magnesium bromide (0.091 g, 0.494 mmol), sealed and heated to 50 °C. After 10 minutes the mixture was cooled and partitioned between dichloromethane and hydrochloric acid (0.2 M aqueous). The organic layer was separated and the aqueous extracted again with dichloromethane. The combined organic layers were dried over sodium sulfate, filtered and concentrated. Preparative HPLC purification (30-70% acetonitrile: water, 0.1% TFA) afforded Compound 3 in enantioenriched form. Chiral HPLC retention time = 6.51 minutes (Chiralpak AS-H, 150 x 4.6 mm, 1 mL/min EtOH). LCMS-ESI* (m/z): [M+H]+ calculated for C21H20F2N3O5 : 432.14; found: 432.2. !H- M (400 MHz, DMSO-d6) δ 12.45 (br s, I H), 10.35 ( i. ./ 5.8 Hz, 1 II), 8.44 (s, H I ). 7.37 (dd, J = 15.2, 8.4 Hz, I H), 7.23 (m, I H), 7.05 (dt, J = 1.8 Hz, 8.7 Hz, IH), 5.44 (dd, J = 9.6, 4.0 Hz), 5.09 (br s, HI), 4.68 (dd, J = 12.8, 4.0 Hz, I H), 4.59 (br s, HI), 4.53 (m, 2H), 4.02 (dd, J = 12.6, 9.4 Hz, IH), 1 .93 (br s, 4H), 1.83 (d, J = 12.4 Hz, I H), 1.57 (m, I I I ).
Example 4
Preparation of Compound 4
(lS,4R)-N-(2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H- l ,4-methanopyrido[r,2':4,5]pyrazino[l ,2-a]pyrimidine-9-carboxamide
Figure imgf000049_0001
Methyl 5-(2,4-difiuorobenzy lcarbamoyl)- 1 -(2,2-dihydroxyeth y 1)~3- methox -4-oxo- 1 ,4~dihydropyridine-2-carboxylate (1-C, 0.12 mmol in 0.53 mL of 9: 1 aeetonitrile:acetic acid containing 0.002 mL methanesulfonic acid) was treated with acetonitrile then (R)-pyrrolidin-3-amine (0,032 mL, 0.36 mmol). The reaction mixture wras capped and heated to 90 °C for 5.5 hours. After cooling, the mixture wras partitioned between dichloromethane and sodium bicarbonate (1M aqueous). The organic layer was separated and the aqueous was extracted again with ethyl acetate. The combined organic layers were dried over sodium sulfate (anhydrous), filtered and concentrated. The residue was dissolved in acetonitrile (1 mL), treated with magnesium bromide (0,022 g, 0.12 mmol), capped and heated to 50 °C for 10 minutes. After cooling the mixture was partitioned between dichloromethane and ammonium chloride (sat). The organic layer was separated and the aqueous was extracted again with dichloromethane. The aqueous layer was adjusted to pH = 1 with HC1 (aq) and extracted again with dichloromethane. The aqueous solution was adjusted to pH = 3 with NaOH (aq) and extracted again with dichloromethane. The combined organic layers were dried over sodium sulfate, filtered, and concentrated. Preparative HPLC purification (10-55% acetonitrile:water, 0.1% TFA) afforded Compound 4. iH-NM (400 MHz, CD3OD-d4) δ 8.42 (s, ! H), 7.42, (q, ./ 7.7 Hz, I H), 6.99 - 6.90 (m, 2H), 5.07 (br s, IH), 4.73 (br d, ./ 10.8 Hz, 1 H), 4.62 (s, 2H), 4.51 (br d, J 12.8 Hz, 1 H), 4.07 (t, J = 1 1.8 Hz, 1H), 3.4- 3.0 (m, 3H), 2.76 (br d, J = 8.8 Hz, I H), 2.15-2.0 (m, 1H), 1 .9-1.8 (m, IH). LCMS-ESI+ (m/z) [M+H calculated for (γ.Η ,.,Γ Λ ,Ο ,: 417.14; found: 17.2,
Example 5
Preparation of Compound 5
(4R., 12aS)-N-(l-(2,4-difluoropheny].)cyclopropyl)-7-hydroxy-4-methyi-6,8-dioxo- 3,4,6,8, 12, i2a-hexahydro-2H-[l ,3]oxazino[3,2-d]pyrido[l ,2-a]pyrazine-9-carboxamide
Figure imgf000050_0001
Figure imgf000051_0001
Step 1
(4R,12aS)-7-methoxy-4-methyl-6,8-dioxo-3 ,4,6,8, 12, 12a-hexahydro- 2H-[l ,3]oxazino[3,2-d]pyrido[ l ,2-a]pyrazine-9-carboxylic acid (Intermediate 5- A) was prepared in an analogous manner to (3S,l laR)-6-methoxy-3-methyl-5,7-dioxo- 2,3,5,7, 1 1 ,1 la-hexahydrooxazolo[3,2-d]pyrido[l ,2-a]pyrazine-8-carboxylic acid as described in WO201 1/1 19566, substituting (R)-3-aminobutan~l~ol for (S)-2~ aminopropan-l-ol . WO20.i l 1 19566 is incorporated herein by reference in its entirety. A suspension of Intermediate 5-A (24.8 mg, 0.080 mmol ), l-(2,4- difluoropheny].)cyclopropanamine HC1 salt (5-15, 21.9 mg, 0.107 mmol), and HATU (48 mg, 0.126 mmol) in CH2C12 (2 mL) was stirred at ambient temperature as N,N- diisopropylethyl amine (DIPEA) (0.1 mL, 0.574 mmol) was added. After 30 minutes, the reaction mixture was diluted with ethyl acetate before washing with 10% aqueous citric acid solution (xl) and saturated aqueous NaHC03 solution (xl). After the aqueous fractions were extracted with ethyl acetate (x l), the organic fractions were combined, dried (MgS04), and concentrated. The residue was purified by combiflash (.12 g column) using hexanes, ethyl acetate, and 20% methanol in ethyl acetate to obtain (4R.,12aS)-N-(l-(2,4-difluoropheny].)cyclopropyl)-7-methoxy-4-methyi-6,8-dioxo- 3,4,6,8, 12, 12a-hexahydro-2H-[ l ,3]oxazino[3,2-d]pyrido[ l ,2-a]pyrazine-9-carboxamide, Intermediate 5-C. LCMS-ESI+ i n/z): | VI - H i calculated for C. l l.. ,!: >VO«: 460.17; found 460.2. Step 2
A suspension of Intermediate 5-C (39 mg, 0.080 mmol) and magnesium bromide (42 mg, 0.2282 mmol) in acetonitri!e (2 mL) was stirred at 50 °C. After 1 hour, the reaction mixture was stirred at 0 °C bath when 1 N BCi (2 mL) was added. After the resulting mixture was diluted with water (~20 mL), the product was extracted with dichloromethane (x3) and the combined extracts were dried (MgS04) and concentrated. The residue was purified by preparative HPLC to obtain (4R,12aS)-N-(l-(2,4- difluorophenyl)cyclopropyl)-7-hydroxy-4-m
2H~[l ,3]oxazino[3,2-d]pyrido[l ,2-a]pyrazine-9-carboxamide, compound 5, as TFA salt. Ή-NMR (400 MHz, C!)Ci„< δ 10.72 (br s, 1 H), 8.37 (s, 1 H), 7.57 (d, J = 7.9 Hz, i l l ). 6.71-6.81 (m, 2H), 5.23 (dd, J = 5.6 and 4.4 Hz, I H), 4.98 (br quint, J = -6.5 Hz, I H), 4.26 (dd, J = 13.6 and 4.4 Hz, IH), 4.12 (dd, J = 13.6 and 5.6 Hz, IH), 4.00-4.06 (m, 2H), 2.16-2.25 (m, IH), 1.55 (br dd, J = 13.8 and 1.8 Hz, IH), 1.40 (d, J = 6.8 Hz, 3H), 1.22-1.31 (m, 4H). 19F NMR (376.1 MHz, CDC13) 8 -76.38 (s, 3F), -1 1 1.69 ~ -1 1 1.645 C m. 2F), LCMS-ESI+ ( n/z): [M+H]"1" calculated for ί ,,H \-F ^O<: 446.15; found: 446.2.
Example 6
Preparation of Compound 6
(l R,4S)-N-(2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-3,4,6,8, 12,12a-hexahydro-2H- 1 ,4-methanopyrido[ 1 ',2':4,5]pyrazino[ 1 ,2-a]pyrimidine-9-carboxamide
Figure imgf000052_0001
6
Methyl 5-(2,4-difluorobenzylcarbamoyl)- 1 -(2,2-dihydroxyethyl)-3- methoxy-4-oxo-l ,4-dmydropyridme~2-carboxyiate (1~C, 0.100 g, 0.243 mmol), (S)~ pyrrolidin-3 -amine (0.043 mL, 0.485 mmol) and potassium carbonate (0.067 g, 0.485 mmol) were suspended in acetonitrile (1.9 mL) and acetic acid (0.1 mL) and heated to 90 °C for 1.5 hours. After cooling, the mixture was treated with magnesium bromide (0.090 g) and heated to 50 °C for 30 minutes. After cooling, the mixture partitioned between dichioromethane and 0.2 M HQ. The organic layer was separated and the aqueous was extracted again with dichioromethane. The combined organic layers were dried over sodium sulfate (anhydrous), filtered and concentrated. Preparative HPLC purification (25-50% acetonitrile:water, 0.1% TFA) afforded Compound 6. 1H-NMR (400 MHz, DMSO-t¾) δ 10.33 (t, J = 6.0 Hz, 1H), 8.44 (s, 1H), 7.48 - 7.32 (m, 1H), 7.31 - 7.15 (m, 1H), 7.14 - 6.97 (m, 1H), 4.86 (d, J= 2.9 Hz, 1H), 4.62 - 4.54 (m, 1H), 4.52 (d, J= 5.9 Hz, 1 H), 4.01 id. J 13.0 Hz, H I ). 2.99 - 2.76 (m, 3H), 1.96 - 1.81 (m, 1H), 1.71 - 1.53 (m, 1H). LCMS-ESI* (m/z): [M+H]'1' calculated for C20H19F2 4O4 : 417.14; found: 417.2.
Example 7
Preparation of Compound 7
(2S,6R)-N-(2,4-difiuorobenzyi)~9 ^
2H-2,6-methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b] [ 1 ,3]oxazocine- 11 -carboxamide
Figure imgf000053_0001
Methyl 5-(2,4-difluorobenzy lcarbamoyl)- 1 -(2,2-dihydroxyethy i)-3- metboxy-4-oxo-l,4-dibydropyridine-2-carboxylate (l-C, 0,050 g, 0.121 mmol), (lS,3R)-3-aminocyclohexanol (0,028 g, 0.243 mmol) and potassium carbonate (0.034 g, 0.243 mmol) were suspended in acetonitrile (0.95 mL) and heated to 90 °C for 0.5 hour. After cooling, acetic acid (0.050 mL) was added and the mixture was reheated to 90 °C for 2h. After cooling the mixture was treated with magnesium bromide (0.044 g) and heated to 50 °C for 1 hour. After cooling, a second portion of magnesium bromide (0.044 g) was added and the mixture was reheated to 50 °C for 15 minutes. After cooling, the mixture partitioned between dichioromethane and 0.2 M HC1, The organic layer was separated and the aqueous was extracted again with dichioromethane. The combined organic layers were dried over sodium sulfate (anhydrous), filtered and concentrated. Preparative HPLC purification (40-80% acetonitrile: water, 0.1% TFA) afforded Compound 7. 'll-NVIR (400 MHz, DMSO- ) δ 12.40 (s, III . 10.36 (t, J = 6.1 Hz, IH), 8.45 (s, Hi).7.48 - 7.29 (m, 1H), 7.31 - 7.13 (m, 1H), 7.13 - 6.97 (m, IH), 5.56 (dd, J = 10.0, 4.1 Hz, III}.4.70 (dd, J = 12.7, 4.1 Hz, 1H), 4.52 (d, J = 5.5 Hz, 2H), 4.40 - 4.29 (m, 2H), 4.06 (dd, J = 12.5, 10.2 Hz, 1H), 2.46 - 2.36 (m, 1H), 1.98 - 1.63 (m, 4H), 1.57 - 1.30 (m, 3H). LCMS~ESI+ (m/z): [M+H]+ calculated for C .H, ·!: .- \ :(),: 446.15; found: 446.2.
Example 8
Preparation of Compound 8
(2R,6S)-N-(2,4-difluorobenzyl)-9-hydroxy-8,l O-dioxo-3,4,5,6,8,1 G,14,14a-oetahydro- 2H-2,6-methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b] [ 1 ,3]oxazoeine- 11 -carboxamide
Figure imgf000054_0001
8
Compound 8 was prepared in a similar manner to compound 7 using (lR,3S)-3-aminocyclohexanol in place of (lS,3R)-3-aminocyclohexanol. !H-NM (400 MHz, DMSO-ii6) δ 12.40 (s, IH), 10.36 (t, J= 6.1 Hz, 1H), 8.45 (s, 1H), 7.48 - 7.30 (rn, IH), 7.23 (td, J ------ 10.6, 2.7 Hz, III), 7.05 (td, J -- 8.3, 2.3 Hz, 1H), 5.56 (dd, J
----- 10.1, 4.1 Hz, IH), 4.70 (dd, J= 12.8, 3.9 Hz, IH), 4.52 (d, J ------- 5.6 Hz, 211).4.39 -
4.27 (m, 21 IK 4.06 (dd, J = 12.6, 10.0 Hz, II IK 2.47 - 2.35 (m, IH), 2.00 - 1.64 (rn. 4H), 1.58 -- 1.30 (m, 3H). LCMS-ESI+ (πι/ζ): \ \ 1 -Hi calculated for C22H22F2 3O5: 446.15; found: 446.2.
Examples 9 and 10
Preparation of Compounds 9 and 1Θ
(2S,5R,13aS)-N-((R)-H4-fluoroph^
octahydro-2,5-methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b][ 1 ,3]oxazepine- 10-carboxamide 9 and (2R,5S,13aR)-N-((R)-l-(4-fj.uorophenyl)ethyl)-8-hydroxy-7,9-dioxo- 2,3,4,5 J,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,
b] [ 1 ,3]oxazepine- 10-carboxamide 10
Figure imgf000055_0001
Figure imgf000055_0002
Step 1
l-(2,2-dimethoxyethyl)-5-methoxy-6-(methoxycarbonyl)-4-oxo-l ,4- dihydropyridine-3-carboxylic acid (1-A, 0.500 g, 1.59 mmol), was suspended in acetomtrile (6 rnL) and treated with N,N-diisopropylethylamine (DIPEA) (0.550 mL, 3.17 nifflol), (R)-l-(4-fluoropbenyi)ethanamine (0.242 rng, 1.74 mmol) and HATXJ (0.661 g, 1.74 mmol). The reaction mixture was stirred for 2 hours and partitioned between ethyl acetate and water. The organic layer was separated and washed with HCl ( 10% aq), sodium bicarbonate (1 M aq), dried over sodium sulfate, filtered and concentrated to afford crude (R)-methyl l -(2,2-dimethoxyethyl)-5-(l -(4- fiuorophenyl)ethylcarbamoyl)-3-methoxy-4-oxo-l ,4-dihydropyridine-2-carboxylate which was used without purification in the next step: LCMS-ESI+ (m/z): [M+H] ; calculated for C" .< ; i ! ^! X -()··: 437.17; found: 437.1.
Step 2
(R)-methyi 1 -(2,2-dimethoxyethyl)-5-(l ~(4- fluorophenyl)ethylcarbamoyl)-3-methoxy-4-oxo- l ,4-dihydropyridine-2-carboxylate was suspended in acetonitrile (5.7 mL) and acetic acid (0.6 raL) and treated with methane sulfonic acid (0.031 mL, 0.477 mmol). The mixture was capped and heated to 75 °C. After 7h, the mixture was cooled and used without purification in the next step: LCMS-ES (m/z): i VI · 1 1 1 calculated for { h;i i■ !- X ( )-: 409.14; found: 409.0. Step 3
(R)-niethyl 1 -(2,2-di ydroxyethyl)-5-(l -(4- fluorophmyl)ethyicarbamoyl)-3-methoxy^-oxo-l ,4-dihydropyridine-2-carboxylate (3.6 mL of the crude mixture from Step 2, 0.8 mmol) was diluted with acetonitrile (3.6 mL) and treated with c s-3-aminocyclpentanoi, HQ salt (0.219 g, 1 ,6 mmol) and potassium carbonate (0.276 g, 2.0 mmol ). The mixture was capped and heated to 90 °C. After 20 minutes, the reaction mixture was cooled and partitioned between dichloromethane and HQ (0.2 M aq). The layers were separated and the aqueous layer was extracted again with dichloromethane. The combined organic layers were treated with a small amount of acetonitrile, dried over sodium sulfate, filtered and concentrated.
The residue was suspended in acetonitrile (4 mL) and treated with magnesium bromide (0.177 g). The mixture was capped and heated to 50 °C. After 10 minutes, the reaction mixture was cooled and partitioned between dichloromethane and HQ (0.2 M aq). The layers were separated and the aqueous layer was extracted again with dichlormethane. The combined organic layers were dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography on silica gel (0-8% ethanol :DCM) to afford a diastereomeric mixture of desired 9 and 10.
The mixture was separated by chiral HPLC using Chiralpak AD-H with 100% ethanol as eluent to afford Compounds 9 and 10 in enantiomericaily enriched form:
For Compound 9: LCMS-ESI+ (m/z): [M+H]+ calculated for C22H23FN3O5: 428.16; found: 428.1. Chiral HPLC retention time = 10.177 minutes (Chiralpak AD-H, 150 x 4.6 mm, 1 mL/min EtOH). 1H- MR (400 MHz, DMSO-<¾) δ 12.45 (s, 1H), 10.45 (d, J = 7.7 Hz, 1H), 8.40 (s, 1H), 7.37 (dd, J = 8.6, 5.6 Hz, 2H), 7.15 (t, J = 8.9 Hz, 2H), 5.44 (dd, J = 9.5, 4.2 Hz, I H), 5.17 - 5.04 (m, 2H), 4.73 - 4.62 (m, I H), 4.59 (s, I H), 4.00 (dd, J = 12.7, 9.5 Hz, 1H), 1 .93 (s, 4H), 1 .83 (d, J = 11.8 Hz, I H), 1.56 (dt ./ 12.1, 3.4 Hz, I H), 1.44 id, J 6.9 Hz, 3H).
For Compound 10: LCMS-ESI+ (w z): i VI · 1 1 1 calculated for C22H23FN3O5: 428,16; found: 428.1. Chiral OPLC retention time = 14.061 minutes (Chiralpak AD-H, 150 x 4.6 mm, 1 mL/min EtOH). 1H-NMR (400 MHz, DMSO-rfe) δ 12.44 (s, IH), 10.46 (d, J = 7.8 Hz, IH), 8.41 (s, IH), 7.37 (dd, J = 8.6, 5.6 Hz, 2H), 7.15 (t, J= 8.9 Hz, 2H), 5.42 (dd, J = 9.6, 4.1 Hz, IH), 5.18 - 5.02 (m, 2H), 4.67 (dd, J - 12.8, 4.2 Hz, IH), 4.59 (s, I H), 4.02 (dd. ./ 12.7, 9.6 Hz, IH), 1.93 (s, 4H), 1.83 (d, J= 12.0 Hz, I H), 1.57 (di. J 13.0, 3.5 Hz, IH), 1.44 (d. ./ 6.9 Hz, 3H). Example 11
Preparation of Compound 11
(2S,5R, 13aS)-N-((R)- 1 -(2,4-difluorophenyl)ethyl)-8-hydroxy-7,9-dioxo-
2,3,4,5,7,9,13,13a-octaliydro-2,5-methanopyrido[1^2':4,5]pyraziiio[2,l- b] [ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000057_0001
Figure imgf000058_0001
Step 1
l -(2,2-dimethoxyethyi)-5-methoxy-6-(methoxycarbonyl)-4-oxo-l,4- dihydropyridine-3-carboxylic acid (1-A, 0.315 g, 1.00 mmol), was suspended in acetonitrile (4 mL) and treated with Ν,Ν-diisopropylethyiamme (DIPEA) (0.348 mL, 2.00 mmol), (R)-l-(2,4-difluorophenyl)ethanamine HC1 salt (0.213 mg, 1.10 mmol) and HATU (0.418 g, 1.10 mmol). The reaction mixture was stirred for 1 hour and partitioned between dichloromethane and FICI (10% aq). The organic layer was separated and washed sodium bicarbonate (1M aq), dried over sodium sulfate, filtered and concentrated to afford crude (R)-methyl 5-(l-(2,4-difiuorophenyl)ethyicarbamoyl )- 1 -(2,2-dimethoxyethyl)-3-methoxy-4-oxo- 1 ,4-dihydropyriditie-2-carboxylate which was used without purification in the next step. LCMS-ESF (m/z): [M+Hy calculated for C21H25F7N2O7: 455.16; found: 455.1.
Step 2
(R)-methyl. 5 -(1 -(2, 4-difluorophenyl)ethyl carbamoyl)- 1 ~(2,2~ dimethoxyethyl)-3-methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate was suspended in acetonitrile (3.6 mL) and acetic acid (0.4 mL) and treated with methane sulfonic acid (0.020 mL). The mixture was capped and heated to 75 °C. After 16 hours, the crude mixture was cooled and used without purification in the next step. LCMS-ES (m/z): [M+H calculated for
Figure imgf000058_0002
427.13; found: 427.1.
Step 3
(R)-methyl 5-(l~(2,4~dii"luorophenyl)ethylcarbamoyl)-l-(2,2- dihydroxyethyl)-3-methoxy-4-oxo-l,4-dihydropyridine-2-carboxyiate (half of the crude mixture from Step 2, approx 0,5 mrnol) was diluted with acetonitrile (2.5 ml.) and treated with (18,3R)-3-aminocyelopentanol (0.110 g, 1.09 rnmol) and potassium carbonate (0.069 g, 0.50 mrnol). The mixture was capped and heated to 90 °C. After 15 minutes, the reaction mixture was cooled and magnesium bromide (0.1 84 g) was added. The reaction mixture was heated to 50 °C. After 10 minutes, the mixture was cooled and treated with an additional portion of magnesium bromide (0.184 g). The reaction mixture was reheated to 50 °C and stirred for 10 minutes. After cooling, the mixture was partitioned between dichloromethane and HC1 (0.2 M aq). The layers were separated and the aqueous layer was extracted again with dichloromethane. The combined organic layers were dried over sodium sulfate, filtered and concentrated. Preparative HPLC purification (30-60% aceionitri le:water, 0.1% TFA ) afforded desired Compound 11. LCMS-ESI+ i n/z): [ V5 l ! j calculated for t . . 1 ί .,Γ ,Ο - 446.15; found: 446.1. 1H- MR (400 MHz, DMSO-afc) δ 12.46 (s, 1 H), 10.53 (d, J = 7.5 Hz, 1H), 8.38 (s, 1H), 7.39 (q, J = 8.5 Hz, 1H), 7.29 - 7.12 (m, 1H), 7.13 - 6.93 (m, 1H), 5.44 (dd, J = 9.8, 4.2 Hz, 1H), 5.28 (p, J = 7.3, 6.8 Hz, 1H), 5.09 (s, 1H), 4.66 (dd, J = 13.2, 4.3 Hz, IH), 4.59 (s, 1H), 3.99 (dd. J 13.1 , 9.6 Hz, I H), 1 .93 (s, 41 1 ), 1.83 (d, J = 12.4 Hz, I H), 1.56 ( di. J 12.5, 2.9 Hz, I H), 1.45 (d. ./ 6.9 Hz, 3H).
Example 12
Preparation of Compound 12
(2R,5S,13aR)-N (R) -(2,4-difluorophenyl)ethyl)-8-hydroxy-7,9-dioxo-
2,3,4,5,7,9, 13, 13a-octahydro-2,5-methanopyrido[ 1 ',2'i4,5]pyrazino[2, 1 - b] [ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000059_0001
12 Compound 12 was prepared in a similar manner to compound 11 using
(l R,3S)-3-ammocyclopentariol in place of (1 S,3R)-3-aminocyclopentanol. 'H- R (400 MHz, DMSCMe) δ 12,43 (s, I H), 10.52 (cL J 8.2 Hz, IH), 8.38 (s, I H), 7.39 (q, ./ = 8.4 Hz, IH), 7.28 - 7.12 (m, 1H), 7.11 - 6.97 (m, IH), 5.41 (del, J = 10.0, 4.0 Hz, IH), 5.35 - 5.20 (m, 1H), 5.08 (s, 1H), 4.65 (ύά. J 13.1, 3.8 Hz, !!!}.4.58 (s, IH), 4.01 icicl../ 12.8, 9.5 Hz, IH), 1.92 (s, 4H), 1.83 id. J 11.5 Hz, III).1.61 - 1.51 (m, 1H), 1.44 (d, J ==: 6.9 Hz, 3H). LCMS-ESI+ (m/z) (VI Hi calculated for C22H22F2N3O5: 446.15; found: 446.1.
Preparation of Compound 13
(2S,5R,13aS)-N-((S)-l-(2,4-difluorophenyl)ethyl)-8-hydroxy-7,9-dioxo-
2,3,4,5,7,9, 13,13a-octahydro-2,5-methanopyrido[ I ',2':4,5]pyrazino[2, 1 - b][l ,3]oxazepine-10-carboxamide
Figure imgf000060_0001
13
Compound 13 was prepared in a similar manner to compound 11 using (S)- 1 -(2,4-difluorophenyl)ethanaminein place of (R)- 1 -(2,4-difluorophenyi)ethanamine, and using only a single portion of magnesium bromide (0.184 g). !H-NM (400 MHz, DMSO-£¾) δ 12.44 (s, IH), 10.53 (d, J= 7.8 Hz, IH), 8.39 (s, IH), 7.39 (q, J= 8.5 Hz, IH), 7.32 - 7.14 (m, IH), 7.05 (t, J ------ 9.1 Hz, IH), 5.42 (dd, J= 9.5, 4.2 Hz, IH), 5.29
(p, J ------ 6.9 Hz, IH), 5.09 (s, IH), 4.65 (dd, J= 12.9, 4.3 Hz, IH), 4.59 (s, IH), 4.02 (dd,
./ 12.6, 9.8 Hz, IH), 1.92 (s, 4H), 1.83 id. J 12.1 Hz, IH), 1.61 - 1.52 (m, IH), 1.44 (d, J = 6.9 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+ calculated for C22H22F2N3O5: 446.15; found: 446.2.
Example 14
Preparation of Compound 14
(2R,5S,13aR)-N-((S)-l-(2,4-dif1uorophenyl)ethyi)-8-hydroxy-7,9-dioxo- 2,3,4,5,7,9, 13,13a-octahydro-2,5-methanopyrido[ I ',2':4,5]pyrazino[2, 1 - b][l ,3]oxazepine-10-carboxamide
Figure imgf000061_0001
14
Compound 14 was prepared in a similar manner to compound 11 using (S)- 1 -(2,4-difl.uorophenyl)ethanamine in place of (R)~ 1 -(2,4-difluorophenyl)ethanamme and using (lR,3S)~3-ammocyclopentanol in place of (lS,3R)-3-aminocyc3.opentanol. 'H-NMR (400 MHz, I ) VI S< >-</„) δ 12.46 (s, 1H), 10.53 (cL 7.6 Hz, III}.8.38 (s, IH), 7.39 (q, ./ 8.6 Hz, ill).7.28 - 7.14 (m. III}.7.05 (t, J ------- 8.5 Hz, Hi).5.44 (dd. J
= 9.8, 3.8 Hz, 1H), 5.28 (p, J= 8.0 Hz, IH), 5.09 (s, 1H), 4.66 (dd, J= 12.9, 4.0 Hz, 1H), 4.59 (s, IH), 3.99 (dd, J = 12.5, 9.6 Hz, IH), 1.93 (s, 4H), 1.83 (d, J= 12.6 Hz, IH), 1.56 (dt J= 13.0, 3.3 Hz, IH), 1.45 (d, J = 6.9 Hz, 3H). LCMS-ESI* (ni/z): [M+H]+ calculated for C22H22F2 3O5: 446.15; found: 446.1.
Example 15
Preparation of Compound 15
(2S,5R,13aS)-N-(4-fluorobenzyl)^
2,5-methanopyrido[ 1 ',2':4,5]pyrazmo[2, 1 -b] [ 1 ,3]oxazepine-l 0-carboxamide
Figure imgf000061_0002
15
Figure imgf000062_0001
Step 1
l -(2,2-dimethoxyethyi)-5-methoxy-6-(methoxycarbonyl)-4-oxo-l ,4- dihydropyridine-3~carboxylic acid (1-A, 3.15 g, 10.0 mmoi), suspended in acetonitri!e (36 mL) and acetic acid (4 mL) was treated with methane sulfonic acid (0.195 mL). The mixture heated to 75 °C. After 7 hours, the crude mixture was cooled and stored in a - 10°C for three days. The crude mixture was reheated to 75 °C for 2 hours, cooled used without purification in the next step. LCMS-ESI+ (m/z): j M I I I calculated for C19H21F2N2O7: 288.07; found: 288.1. Step 2
Grade l-(2,2-dihydroxyethyl)-5-methoxy-6-(methoxycarbonyl)-4-oxo- i ,4-dihydropyridine-3-carboxylic acid (16.8 mL of crude mixture from Step 1 , approx 4 mmol) was combined with (l S,3R)-3-aminocyclopentanol (0.809 g, 8 mmol), diluted with acetoiiitrile (16.8 mL), and treated with potassium carbonate (0.553 g, 4 mmol). The reaction mixture was heated to 85 °C, stirred for 15 minutes, cooled to ambient temperature and stirred an additional 16 hours. HCl (50 mL, 0.2M aq) was added and the clear yellow solution was extracted three times with dichloromethane. The combined organics were dried over sodium sulfate, fi ltered and concentrated to a yellow solid. This crude material was precipitated from dichloromethane/hexanes to afford desired intermediate 15-B as a light beige powder. 1H-NMR (400 MHz, DMSO- d6) δ 8.72 (s, 1H), 5.42 (dd, J = 9.6, 4.1 Hz, 1H), 5.09 (s, 1H), 4.72 (dd, J = 13.0, 3.7 Hz, 1 H), 4.57 (s, 1 H), 4.09 (dd, ./ 12.5, 9.6 Hz, H I ). 3.83 (s, 3H), 1.92 (s, 3H), 1 .78 Cm. 2H), 1.62 - 1.47 (m, ! ! ! }. LCMS-ESI+ (m/z): [M+Hj calculated for (', Ι , -Ν .(>„:
321.1 1 ; found: 321.2.
Step 3
Intermediate 15-B (0.040 g, 0.125 mmol) and (4- fluoroplienyljmethaiiamine (0.017 g, 0.137 mmol) were suspended in acetomtriie (1 mL) and treated with Ν,Ν-diisopropylethylamine (DIPEA) (0.033 mL, 0.187 mmol) and HA.TU (0.052 g, 0.137 mmol). After stirring for 30 minutes, the reaction mixture was treated with magnesium bromide (0.046 g, 0.25 mmol) and heated to 50 °C. After 10 minutes, the reaction mixture was cooled and treated with HC1 (2 mL, 10% aq). After a fewr minutes, the precipitate was filtered and washed with HC1 (10% aq) and water. Preparative HPLC purification of the precipitate (20-65% aeetonitrilerwater, 0.1% TFA) afforded desired Compound 15. 3 H-NMR (400 MHz, DMSO-</6) δ 12.44 (s, 1H), 10.36 (t, J = 6.0 Hz, 1H), 8.46 (s, 1H), 7.37 - 7.28 (m, 2H), 7.19 - 7.09 (m, 2H), 5.43 (dd, J = 9.6, 4.0 Hz, 1H), 5.08 (s, 1H), 4.68 (dd, J = 12.8, 4.1 Hz, 1H), 4.59 (s, ! H), 4.58 - 4.42 (m, 3H), 4.02 (dd, J = 12.7, 9.6 Hz, I I I . 1.92 (s, 5H), 1.83 (d, J = 12.2 Hz, 1 H), 1.56 < dl, ./ 12.0, 3.4 Hz, 1 H). LCMS-ES1+ (m/z): [M+H]'4 calculated for C21H21FN3()5 : 414.15; found: 414.2.
Example 1
Preparation of Compound 1
(2S,5R,13aS)-N-(2,3-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7
octahydro-2,5-meth nopyri.do[1 ^2':4,5]pyrazino[2J.-bj[l , 3joxazepme-10-carboxami.de
Figure imgf000063_0001
16
Compound 16 was prepared in a similar manner to compound 15 using (2,3-difluoropheny].)methanamine in place of (4-fiuorophenyi)methanamine, 1H-NMR (400 MHz, DMSO- ) δ 12.46 (s, i l l ). 10.41 (t, J = 6.1 Hz, 1H), 8.45 (s, I I I }. 7.43 - 7.25 (m, i l l ). 7.25 - 7.05 (m, 2H), 5.44 (dd, ./ 9.5, 3.9 Hz, 1H), 5.09 (s, 1H), 4.68 (dd, ./ = 12.8, 4.0 Hz, 1H), 4.65 - 4.53 (m, 3H), 4.02 (dd, ./ = 12.7, 9.8 Hz, 1H), 3.56 (s, 1H), 1 .93 (s, 4H), 1.83 (d, ./ 1 1.9 Hz, 1H), 1.57 (dt, ./ 1 1.5, 3.0 Hz, ! H). LCMS-ESI* (m/z): [M+H]+ calculated for C21H20F2N3O5 : 432.14; found: 432.2. Example 17
Preparation of Compound 17
(2S,5R43aS)-N 4-chloro-2-fluorobmzyi)-8½droxy-7,9-dioxo-2,3,4,5,7,9,13,13a- octahydro-2,5-meth nopyri.do[l ^2':4,5]pyrazino[2 J. -bj[l , 3joxazepme-l.0-carboxami.de
Figure imgf000064_0001
17
Compound 17 was prepared in a simi lar manner to compound 15 using (4-chloro-2-fluorophenyl)methanamine in place of (4-fiuorophenyi)methanamine. iH- NMR (400 MHz, DMSC 6) δ 12.46 (s, 1H), 10.45 - 10.29 (m, 1 H), 8.44 (s, 1H), 7.42 (dd. 10.0, 2.0 Hz, 1H), 7.33 (t, J = 8.1 Hz, 1H), 7.26 (dd, J = 8.4, 1.8 Hz, 1H), 5.50 - 5.38 (m, 1H), 5.09 (s, 1H), 4.68 (dd, J = 13.0, 4.0 Hz, 1H), 4.59 (s, 1H), 4.54 (m, 2H), 4.02 (dd, J = 12.8, 9.7 Hz, 1H), 1.93 (s, 4H), 1.83 (d, J = 12.0 Hz, 1H), 1.57 (dt, J = 1 1.9, 3.4 Hz, 1 H). LCMS-ESI"' (m/z) [M+Hj+ calculated for C21H20CIFN3O5: 448.1 1 ; found: 448.2.
Preparation of Compound 18
(2S,5R, 13aS)-N-(3,4-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13a- octahydro-2,5-methanopyrido[ 1 ',2':4,5 jpyrazino[2, 1 -b j[ 1 ,3 joxazepine- 10-carboxamide
Figure imgf000065_0001
Compound 18 was prepared in a similar manner to compound 15 using (3,4-dif3.uorophenyl)methanamine in place of (4-fluoropheny{)methanamine. !H- MR (400 MHz, DMSO- ) 6 12.46 (s, IH), 10.51 - 10.27 (m, I H), 8.46 (s, 1H), 7.50 - 7.23 (m, 2H), 7.23 - 7.03 (m, 1 H), 5.44 (dd, ./ 9.5, 3.6 Hz, 1 H), 5.09 (s, 1 H), 4.75 - 4.63 (m, I H), 4.60 (s, 1 1 1 ). 4.57 - 4.44 (m, 2H), 4.02 (dd, J = 12.6, 9.8 Hz, IH), 1.93 (s, 4H), 1.83 (d, J = 12.0 Hz, IH), 1.57 (dt, J = 12.0, 3.4 Hz, IH). LCMS-ESI+ (m/z): [ VI H i calculated for C21H20F2N3O5: 432.14; found: 432.2. Example 19
Preparation of Compound 19
(lR,5S)-N~(2,4~difluorobe^
n ethanopyrido[r,2':4,5]pyrazmo[l ,2-a][l ,3]diazepine-l 0-carboxamide
Figure imgf000065_0002
Steps 1 and 2
Methyl 5-(2,4-difluorobenzylcarbamoyl)- 1 -(2,2-dihydroxyethyl)-3- metiioxy-4-oxo-l ,4-diiiydropyridine-2-carboxyiate (l-C, 97.5 mg, 0.236 mmol) was treated with acetoiiitrile (1.9 mL), acetic acid (0, 1 mL), potassium carbonate (145 mg, 1 .05 fflfflol), and (8)~piperidm-3-amme dihydrochloride (82 rag, 0.472 rnmol). The reaction mixture was sealed and heated to 90 °C. After 60 minutes, the reaction mixture was cooled partitioned between brine and dichloromethane. The aqueous phase was thrice extracted into dichloromethane and the combined organic phases were combined, dried over MgS04, filtered, concentrated. The crude product was dissolved into acetonitrile (2 mL) and magnesium bromide (89.1 mg, 0.48 mmol) was added. The mixture was reseaied and heated to 50 °C. After 90 minutes, the reaction mixture was quenched with ~5 mL of 0.2M HCl(aq), the pH adjusted to -10, diluted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile:water, 0.1% TFA) afforded Compound 19. 'Sl- MR (400 MHz, Chloroform-d) δ 10.43 (t, J = 5.9 Hz, 1 H), 8.43 (s, 1 H), 7.39 - 7.30 (m, 1 H), 6.81 (q, J = 8.1 Hz, 2H), 4.89 (dd, J = 1 1.6, 3.8 I ! . i l l ). 4.69 (s, 1H), 4.64 (d, J = 5.8 Hz, 2H), 4.26 (dd, J = 12.6, 3.8 Hz, 1H), 3.91 (t, J = 12.1 Hz, 1H), 3.20 - 3.10 (m, 2H), 3.06 (s, 21 ! }. 2.14 - 2.02 (m, 1 H), 1.96 - 1 .81 (m, 2H), 1.81 ! .7() (m, 1H). LCMS-ESf (m/z): [M+H]+ calculated for C21H20F2N4O4: 431.15; found: 431.2.
Example 20
Preparation of Compound 20
(l S,5R)-N-(2,4-difiuorobenzy])-8-hydroxy-7,9-dioxo-2,3,4,5,7,9, 13,13a-octahydro-l ,5- methanopyrido[ 1 ',2':4,5]pyrazmo[ 1 ,2-a] [ 1 ,3]diazepine- 10-carboxamide
Figure imgf000066_0001
20
Figure imgf000066_0002
Steps 1 and 2
Methyl 5-(2,4-difluorobenzy lcarbamoyl)- 1 -(2,2-dihydroxyethy i)-3- methoxy-4-oxo- 1 ,4-dihydropyridine-2-carboxyiate (1-C, 103,3 mg, 0.25 mmol) was treated with acetonitrile (1 ,9 mL), acetic acid (0.1 mL), potassium carbonate (159.8 mg, 1.16 mmol), and (R)-piperidin-3 -amine dihydrochloride (90 mg, 0.52 mmol). The reaction mixture was sealed and heated to 90 °C. After 40 minutes, the reaction mixture was cooled partitioned between brine and dichloromethane. The aqueous phase was thrice extracted into dichloromethane and the combined organic phases were combmed, dried over MgS04, filtered, concentrated. The crude product was dissolved into acetonitrile (2 mL) and magnesium bromide (96.5 mg, 0.52 mmol) was added. The mixture was reseated and heated to 50 °C. After 80 minutes, the reaction mixture was quenched with ~5 mL of 0.2M HC1 (aq), the pH adjusted to - 10, diluted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile : water, 0.1% TFA) afforded Compound 20. !H- M (400 MHz, DMSO-d6) 8 10.35 (t, J = 6.0 Hz, 1H), 8.48 (s, 1H), 7.45 - 7.33 (m, 1H), 7.29 - 7.18 (m, 1H), 7.05 (td, J = 8.5, 2.4 Hz, 1H), 5.06 (dd, J = 1 1.4, 3.5 Hz, 1 H), 4.56 - 4 47 (m, M l ). 4.44 (s, 1H), 4.05 (t, J - 1 1 .8 Hz, 1 H), 3.07 - 2.89 (m, 4H), 1.85 - 1 .73 (m, 3H), 1.54 - 1.46 (m, 1H). LC S-ES (m/z): \ \\ i l l calculated for C . ; i [ ·,,] ' . \ id : 431 .15; found: 431.2.
Example 21
Preparation of Compound 21
(2S,5R, 13aS)-N-((S) -(4-fluorophenyl)emyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,n octahydro-2,5-meth nopyri.do[r,2':4,5]pyrazino[2, l-bj[l ,3]oxazepme-10-carboxamide
Figure imgf000067_0001
21
Figure imgf000068_0001
Steps 1 and 2
(S)-Methyl 1 -(2,2-dihydroxyethyi)-5-( 1 -(4- fluorophenyl)ethylcarbamoyl)-3-methoxy-4-oxo- l ,4-dihydropyridine-2-carboxylate (21-A, 1 mL, 0.23 M solution in 19: 1 acetonitriletacetic acid, prepared as per (R)- methyl l-(2,2-dihydroxyethyl)-5-(l-(4-fluorophenyl)ethylcarbamoyl)-3-methoxy-4- oxo-l ,4-dmydropyridine-2-carboxylate 9-A from Example 9 using (S)-l-(4- fluorophenyljethanamine in place of (R)-l-(4-fl.uorophenyl)ethanamme) was treated with (1 S,3R)-3-aminocyclopentanoi (62 mg, 0.61 ramol) and potassium carbonate (34 mg, 0.25 mmoi). The reaction mixture was sealed and heated to 90 °C. After 60 minutes, the reaction mixture was cooled partitioned between brine and dichloromethane. The aqueous phase was thrice extracted into dichloromethane and the combined organic phases were combined, dried over MgS04, filtered, and concentrated. The crude product was dissolved into acetonitrile (2 mL) and magnesium bromide (74 mg, 0.4 mrnol) was added. The mixture was resealed and heated to 50 °C. After 100 minutes, the reaction mixture was quenched with 0.2Λ1 HQ (aq), diluted with brine, and thrice extracted into DCM. H PLC purification (acetomtri leiwater, 0.1% TFA) afforded Compound 21. 1H- MR (400 MHz, D.'viSO -d,.} δ 12.42 (br s, IH), 10.45 (d, J = 7.9 Hz, IH), 8.40 (s, IH), 7.36 (dd, J = 8.6, 5.5 Hz, 2H), 7.14 (t, J = 8.9 Hz, 2H), 5.42 (dd, J = 9.6, 4.2 Hz, IH), 5.15 - 5.04 (m, 2H), 4.72 - 4.55 (m, 2H), 4.02 (dd, J = 12.7, 9.7 Hz, IH), 1.97 - 1.89 (m, 4H), 1.82 (d, J = 12.2 Hz, IH), 1.56 (dt, J = 1 1.9, 3.3 Hz, IH), 1.43 (d, J = 6.9 Hz, 3H). LCMS-ESI+ (m/z): i VI · i i | calculated for C22H22FN3O5: 428.16; found: 428.1.
Example 22
Preparation of Compound 22 (2 R,5 S, 13aR)-N-((S)- 1 -(4-fluorophenyl)ethyl)-8-hydioxy-7,9-dioxo-2.3 ,4,5 ,7,9, 13 , 13a- octahydro-2,5-methanopyrido[ 1 \2':4,5]pyrazmo[2, 1 -b][ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000069_0001
22
Figure imgf000069_0002
Steps 1 and 2
(S)-methyl 1 -(2,2-dihydroxyethy[)-5-(l -(4- fluorop enyl)etbyicarbamoyl)-3-metboxy-4-oxo-l ,4-dibydropyridine-2-carboxylate (21-A, 1 niL, 0.23 M solution in 19: 1 acetoniirile: acetic acid) was treated with (1R.3S)- 3-aminocyclopentanol (52 mg, 0.51 mmol) and potassium carbonate (31 mg, 0.22 mmol). The reaction mixture was sealed and heated to 90 °C. After 60 minutes, the reaction mixture was cooled partitioned between brine and dichioromethane. The aqueous phase was thrice extracted into dichioromethane and the combined organic phases were combined, dried over MgS04, filtered, and concentrated. The crude product was dissolved into acetoniirile (2 mL) and magnesium bromide (91 mg, 0.49 mmol ) was added. The mixture was resealed and heated to 50 °C. After 100 minutes, the reaction mixture was quenched with 0.2M HCl(aq), diluted with brine, and thrice extracted into DCM. HPLC purification (acetoniirile: water, 0.1% TFA) afforded Compound 22. ¾-NM (400 MHz, DMSO-d6) 8 12.44 (br s, 1H), 10.45 (d, J = 7.7 Hz, 1H), 8.39 (s, 1H), 7.36 (dd, J = 8.5, 5.6 Hz, 2H), 7.14 (t, J = 8.9 Hz, 2H), 5.43 (dd, J - 9.6, 4.0 Hz, 1 H), 5.15 - 5.06 (m, 21 1 ), 4.66 (dd, J - 12.8, 3.9 Hz, 1 1 1 ), 4.58 (s, ! H), 3.99 (dd, J = 12,6, 9.5 Hz, i l l ). 1 .93 (s, 41 1 ). 1.82 id. J - 12,0 Hz, i l l ). 1.56 (dt, J 12.0. 3.0 Hz, 1 1 ! ). 1.44 id. J = 6.9 Hz, 3H). LC S-ESI+ (m/z): | V] H i calculated for C22H22FN3O5: 428.16; found: 428.1. Example 23
Preparation of Compound 23
(2S,5 I , 13aS)-N-(2-fl orobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9, 13,13a-octahydro- 2,5-metbanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b] [ 1 ,3]oxazepine-10-carboxamide
Figure imgf000070_0001
Figure imgf000070_0002
Steps 1 and 2
15-B (41 mg, 0.13 mmol) was treated with acetonitrile (1 mL), (2- fluoropbenyl)methanamine (.17 mg, 0.14 mmol), HATU (67 mg, 0.18 mmol), and N,N- diisopropylethylamine (DIPEA) (24 mg, 0.19 mmol). The reaction mixture was stirred at room temperature for one hour and magnesium bromide (47 mg, 0.26 mmol) was added. The mixture was sealed and heated to 50 °C. After 60 minutes, the reaction mixture was quenched with 0.2M HC1 (aq), diluted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile:water, 0.1 % TFA) afforded Compound 23. iH- NMR (400 MHz, Chloroform-d) δ 10.42 (s, IH), 8.34 (s, IH), 7.36 (t, J = 7.9 Hz, IH), 7.24 - 7.17 (m, I H), 7.12 - 6.97 (m, 2H), 5.40 - 5.32 (m, I H), 5.29 (t, J = 3.5 Hz, I H), 4.67 (s, M l ). 4.28 - 4.20 (m, I H), 4.06 - 3.95 (m, I H), 2.20 - 1.96 (m, 4H), 1 .95 - 1.84 (m, IH), 1 .59 (dt, J = 12.4, 3.3 Hz, IH). LCMS-ESF (m/z): j VM i j calculated for C21H20FN3O5: 414.15; found: 414.2. Example 24
Preparation of Compound 24
(2S,5R,13aS)-N-(3,5-diiluorobeiizyl)-8-liydroxy-7,9-dioxo-2,3,4
octahydro-2,5-meth nopyri.do[1 ^2':4,5]pyrazino[2J.-bj[1 ]oxazepme-10-carboxamide
Figure imgf000071_0001
Steps 1 and 2
15-B (44 mg, 0.14 mmol) was treated with aeetonitriie (1 mL), (3,5- dif3.uorophenyl)met anamine (32 mg, 0.23 mmol), HA.TU (54 mg, 0.14 mmol), and N.N-diisopropylemylamine (37 mg, 0.29 mmol). The reaction mixture was stirred at room temperature for one hour and magnesium bromide (57 mg, 0.31 mmol) was added. The mixture was sealed and heated to 50 °C. After 60 minutes, the reaction mixture was quenched with 0.2M HCl (aq), diluted with brine, and thrice extracted into DCfVl HPLC purification (Acetonitri!e:water, 0.1 % TFA) afforded Compound 24. 3"H- NMR (400 MHz, Chlorofonn-d) δ 10.39 (s, 1H), 8.42 (s, 1H), 6.82 (d, J = 7.9 Hz, 2H), 6.65 (t, J = 8.8 Hz, 1H), 5.38 (d, J = 7.7 Hz, 1H), 5.28 (s, 1H), 4.78 - 4.41 (m, 3H), 4.32 (d, J = 12.1 Hz, H I ). 4.02 (t, J - 10.9 Hz, 1H), 2.30 - 1.97 (m, 4H), 1.97 - 1.81 (m, ), 1.59 (d, J = 12.3 Hz, 1H). LCMS-ESI (m/z): [M+H]+ calculated H19F2N3O5 : 432.14; found: 432.2.
Preparation of Compound 25
(2S,5R,13aS)-N-(4-fluoro-3-(1rifluorom.ethyl)benz.yl)-8-hydroxy-7,9-dioxo- 2,3,4,5,7,9,13, 13a-octahydro-2,5-rnethanopyrido[ ,2':4,5]pyrazino[2, 1 - b] [ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000072_0001
Steps 1 and 2
15-B (43 mg, 0.13 mmol) was treated with acetonitrile ( 1 mL), (4-fluoro- 3-(trifluoromethyi)phenyi)metiiaiiamine (29 mg, 0.15 mmol), HATU (62 mg, 0.16 mmol), and N,N-diisopropyIethylamine (26 mg, 0.20 mmol). The reaction mixture was stirred at room temperature for one hour and magnesium bromide (62 mg, 0.34 mmol) was added. The mixture was sealed and heated to 50 °C. After 60 minutes, the reaction mixture was quenched with 0.2M HCl(aq), di luted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile: water, 0.1% TFA) afforded Compoimd 25. 1H~ NMR (400 MHz, Chloroform-d) δ 10.44 (s, 1 H), 8.29 (s, 1H), 7.56 - 7.38 (m, 2H), 7.06 (t, J = 9.2 Hz, 1H), 5.30 (dd, J - 9.3, 3.5 Hz, I I I . 5.21 (s, 1 H), 4.65 - 4.45 (m, 3H), 4.21 (dd, J = 12.8, 3.4 Hz, 1H), 3.95 (dd, J = 12.4, 9.7 Hz, 1H), 2.1 1 - 1.89 (m, 4H), 1.89 - 1.74 (m, 1 H), 1 .53 (dt, J = 12.4, 3.2 Hz, 1H). LCMS-ESI+ (m/z): \ \\ U calculated for C22H19F4N3O5: 482.14; found: 482.2.
Example 26
Preparation of Compound 26
(2S,5R,13aS)-N-(4-chloro-3-fluoroto
octahydro-2,5-meth nopyri.do[1 ^2':4,5]pyrazino[2J.-bj[l ,3joxazepme-l -carboxamide
Figure imgf000073_0001
Steps 1 and 2
15~B (41 mg, 0.13 mmoi) was treated with acetonitrile (1 ml.), (4- chloro-3-fluorophenyl)methanamine (40 mg, 0.25 mmol), HATU (60 mg, 0.16 mmol), and Ν,Ν-diisopropyfethylamine (28 mg, 0.22 mmol). The reaction mixture was stirred at room temperature for one hour and magnesium bromide (48 mg, 0.26 mmol) was added. The mixture was sealed and heated to 50 °C. After 60 minutes, the reaction mixture was quenched with 0.2M HCl (aq), diluted with brine, and thrice extracted into DCM. HPLC purification
Figure imgf000074_0001
0.1% TFA) afforded Compound 26. 1H- NMR (400 MHz, Chloroform-d) δ 10.41 (s, Hi), 8.30 (s, I Hi.7,24 (t, J = 6.1 Hz, !H), 7.13 - 6.90 (m, 2H), 5.30 (dd, J = 9.1, 3.2 Hz, 1H), 5.22 (s, 1H), 4.61 (s, lH), 4.51 (s, 2H), 4.20 (d, J = 9.4 Hz, Hi).3.95 id. J = 12,0 Hz, 1H), 2.11 - 1.90 (m, 4H), 1.90 - 1.76 (m, 1H), 1.53 (d, J = 12.2 Hz, 1H). LCMS-ESI+ ( /z): [M+H] calculated for C21H19CIF 3O5: 448.11; found: 448.2.
Example 27
Preparation of Compound 27
(2S,5R)-N-(l-(2,4-difluoroph.eny[)cyclopropyl)-8-hydroxy-7,9-d.ioxo- 2,3,4, 5,7, 9,13 ,13a-octahydro-2,5-methanopyrido[ ,2':4,51pyrazino[2,l- b] [1 ,3]oxazepme- 10-carboxamide
Figure imgf000074_0002
Step 1
A suspension of the compound 1-A (1.004 g, 3.19 mmol), the amine 27- A (688 mg, 3.35 mmol), and HATU (1.453 g 3.82 mmol) in ( Ί i >( "!■ (20 mL) was stirred in 0 °C bath as Ν,Ν-diisopropylethylamine (DIPEA) ( 2 mL., 1 1.48 mmol) was added. After 1 hour at 0 °C, the reaction mixture was concentrated to a syrup, diluted with ethyl acetate, and washed with water (x 2). After the aqueous fractions were extracted with ethyl acetate (x 1), the organic fractions were combined, dried (Na^SC^), and concentrated. The residue was purified by CombiFlash (120 g column) using hexanes- ethyl acetate as eluents. The major peak was combined and concentrated to afford 1.082 g (73%) of the product 27-B. After the minor peak was combined and concentrated, the concentrated residue was dissolved in CH2CI2 and some insoluble materials were filtered. The filtrate was concentrated to get 361 mg (24%) of the additional product 27-B. LCMS-ESI+ (rn/z): [M+H]+ calculated for C22H25F2N207: 467.16; found: 467.1. Step 2 and 3
Compound 27-B (81 mg, 0.174 mmol) was dissolved in a mixture (1 mL) of acetonitriie (22 mL), AcOH (2 mL), and methanesulfonic acid (0.14 mL, 2.16 mmol) at room temperature and the resulting solution was stirred at 65 °C for 20 hours.
After the resulting solution was cooled to room temperature, the aminoalcohol 27-D (50 mg, racemic, 0.363 mmol ), 2C03 (50 mg, 0.362 mmol), and acetonitriie (2 mL) were added to the solution. The resultmg mixture was stirred at 65 °C bath for 1 hour. After the reaction mixture was cooled to room temperature, it was acidified with 1 N HC1 (~2 mL), diluted with water (~8 mL), and extracted with CH2C12 (x 3). Combined extracts were dried (Na2S04), concentrated, and purified by CombiFlash to obtain 67 mg (82%) of compound 27-E. ' l l-NVI R (400 MHz, CDCI3) δ 10.53 (s, I H), 8.25 (s, H I ). 7.60 (td, ./ - 8.5, 6.5 Hz, ! ! ! }. 6.85 - 6.57 (m, 2H), 5.33 (br, 1 H), 5.26 (dd, ./ 9.6, 3.9 Hz, I I I }. 4.60 (t, J = 3.0 Hz, 1 1 1 ). 4.1 8 - 4.06 (m, IH), 4.01 (s, 3H), 3.92 (dd. 12.7, 9.6 Hz, IH) , 2, 1 1 - 1.91 (m, 4H), 1.88 - 1.71 (rn. i l l ). 1.60 - 1.49 (m, IH), 1.31 - 1. 10 (m, 4H). "F-NMR (376.1 MHz, CDC13) δ -1 1 1.80 (q, J = 8.8 Hz, I F), -1 12.05 (p, J = 7.9 Hz, I F). LCMS-ES1+ (m/z): [M+H]+ calculated for C24H24F2N3O5 : 472.17; found: 472.1.
Step 4
A mixture of compound 27-E (67 mg, 0.142 mmol) and MgBr2 (66 rag, 0.358 mmol) in MeCN (3 mL) was stirred at 50 °C for 30 minutes and cooled to 0 °C before treating with 1 N HCl (3 mL). After the mixture was diluted with water (-30 mL), the product was extracted with CH2C12 (x 3), and the combined extracts were dried (Na2S04) and concentrated. The product was purified by preparative HPLC and freeze-dried to obtain product 27 as a 1 : 1 mixture with trifluoroacetic acid. 1H-NMR. (400 MHz, CDCI3) δ 10.70 (s, IH), 8.35 (s, IH), 7.57 (q, J = 8.2 Hz, 1H), 6.91 - 6.56 (m, 2H), 5.31 (dt, 14.3, 4.0 Hz, 2H), 4.68 (s, I H), 4.22 (dd, J = 13.2, 3.9 Hz, I H), 3.99 (dd, J = 12.8, 9.3 Hz, I H), 2.28 - 1.96 (m, 5H), 1.88 (ddt, J = 12.1 , 8.6, 3.7 Hz, IH), 1 .71 - 1.49 (m, I H), 1.38 - 1.1 1 (m, 4H). 19F-NMR (376.1 MHz, CDCI3) δ -76.37 (s, 3F), -1 1 1.6 ~ -1 1 1.75 (m, 2F). LCMS-ESI+ (m/z): [M+H calculated for C23H22F2N3O5 : 458.15; found: 458.1.
Example 28
Preparation of Compound 28
(2S,6R)-N-(l -(2,4-difluoropheny[)cyclopropy[)-9-hydroxy-8,10-dioxo- 3,4,5,6,8,10, 14,14a-octahydro-2H-2,6-methanopyrido[l 2,:4,5]pyrazino[2, l- b][l ,3]oxazocine- l 1-carboxamide
Figure imgf000076_0001
Figure imgf000077_0001
Step 1 and 2
Compound 27-B (87 mg, 0.187 mmol) was dissolved in a mixture (2 mL) of acetonitriie (22 mL), AcOH (2 mL), and methanesulfonic acid (0.14 mL, 2.16 mmol ) at room temperature and the resulting solution was stirred at 65 C for 20 hours.
After the resulting solution was cooled to room temperature, the aminoalcohol 28-A (44 mg, racemic, 0.382 mmol) and acetonitriie (2 mL) were added to the solution. After the resulting mixture was stirred at 65 °C bath for 30 minutes, K2C03 (41 mg, 0.297 mmol) was added and the mixture was stirred at 65 °C for 21 hours. The reaction mixture was cooled to room temperature, it was acidified with 1 HC1 (~2 mL), diluted with water (~8 mL), and extracted with CH?C1? (x 3). Combined extracts were dried (\';s -SO .}, concentrated, and purified by preparative HPLC and the fraction containing the product was freeze-dried. After the residue was dissolved in ethyl acetate, the solution was washed with saturated NaHCCh (x 1), dried (Na2S04), and concentrated to obtain 18 mg (20%) of compound 28-15 as a 1 : 1 mixture with trifluoroacetic acid. 1H~NMR (400 MHz, CDC13) δ 10.54 (s, 1H), 8.26 (s, IH), 7.63 (td, J= 8.6, 6.6 Hz, 1H), 6.76 (dddd, J = 21.9, 11.2, 8.7, 2.3 Hz, 2H), 5.39 (dd, J = 9.6, 3.7 Hz, IH), 4.53 - 4.36 (m, 2H), 4.09 (dd, 12.8, 3.7 Hz, I H), 4.03 (s, 31 ! ). 3.99 (dd, J = 12.7, 9.7 Hz, IH), 2.41 - 2.20 (m, 2H), 1.84 (did. J 19.7, 9.3, 8.8, 4.4 Hz, 2H), 1.74 (dd, J 14.6, 2.5 Hz, I H), 1 .62 - 1.35 (m, 2H), 1.34 - 1 .14 (m, 5H). "F-NMR (376.1 MHz, CDCL) δ -111.75 (q, J = 8.9 Hz, I F), -1 12.01 (p, J = 7.9 Hz, I F). LCMS-ESI+ (m/z): [M+Hf calculated for <'.>·. I L-.F -.VO-:: 486.18; found: 486.2.
Step 3 Compound 28-B (18 mg, 0.037 mmol) was treated with MgBr2 as described in step4 in the synthesis of compound 27-E to obtain compound 28, 1H- NMR (400 MHz, CDC13) 810.66 (s, 1H)S 8.29 (s, IH), 7.59 (td, J= 8.5, 6.6 Hz, 1H), 6.89 - 6.60 (m, 2H), 5.51 (dd, J= 9.9, 4.0 Hz, ill).4.55 (s, IH), 4.48 (t, ./ 4.2 Hz, IH), 4.21 (dd, J= 12.9, 4.1 Hz, !!!}.3.99 (dd, J = 12.8, 9.8 Hz, 1H), 2.56 - 2.35 (m, lH), 2.14 (dd, J= 16.1, 5.9 Hz, IH), 1.96 - 1.74 (m, 3H), 1.66 - 1.37 (m, 3H), 1.28 (d, ./' 4.4 Hz, 2H), 1.26 - 1.19 (m, 2H). WF-NMR (376.1 MHz, CDC13) δ -76.41 (s, 3F, - 111.79 (m, 2F). LCMS-ESI+ (m/z): [M+H]+ calculated for ( Π..,!:Λ :() 472.17; found: 472.1.
Preparation of Compound 29
(2R.,6S)-N-(l-(2,4-difiuorophenyl.)cyclopropyl.)-9-hydroxy-8,10-dioxo- 3,4,5,6,8, 10, 14, 14a-octahydro-2H-2,6-methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 - b][l,3]oxazocine-l 1-carboxarnide
Figure imgf000078_0001
Figure imgf000078_0002
Step 1 and 2
Compound 29-B (13 mg, 14%) was prepared from compound 27-B (87 mg, 0,187 mmol) and the aminoalcohol 29-A (45 mg, 0.391 mmol) in a manner similar to that described in step 1 of the synthesis of compound 28-B. 1H-NMR (400 MHz, C!X h) δ 10.54 (s, III).8.26 (s, 1H), 7.63 (td, J = 8.6, 6.6 Hz, lH), 6.76 (dddd, J = 21.9, 11.2, 8.7, 2.3 Hz, 2H), 5.39 (dd, J= 9.6, 3.7 Hz, 1H), 4,53 - 4,36 (m, 2H), 4.09 (dd, J= 12.8, 3.7 Hz, lH), 4.03 (s, 3H), 3.99 (dd, J= 12.7, 9.7 Hz, lH), 2.41 - 2.20 (m, 2H), 1.84 (did. J = 19.7, 9.3, 8.8, 4.4 Hz, 2H), 1.74 (dd, J = 14.6, 2.5 Hz, 1H), 1.62 - 1.35 (m, 2H), 1.34 - 1.14 (m, 5H). 19F-NMR (376.1 MHz, CDC1) δ -111.75 (q, J= 8.9 Hz, IF), -112.01 (p, J = 7.9 Hz, IF). LCMS-ESI+ (m/z) [M+H]+ calculated for C2.5H26F2N.3O5: 486.18; found: 486.2.
Step 3
Compound 29 was prepared from compound 29-B in a manner similar to that described in step 2 of the synthesis of compound 16. 1H-NMR (400 MHz, ( DC!:) 610.66 (s, 1H), 8.29 (s, 1H), 7.59 (td, J - 8.5, 6.6 Hz, 1H), 6.89 - 6.60 (m, 2H), 5.51 (dd, J= 9.9, 4.0 Hz, 1H), 4.55 (s, lH), 4.48 (t, J = 4.2 Hz, lH), 4.21 (dd, J = 12.9, 4.1 Hz, III .3.99 (dd, J - 12.8, 9.8 Hz, IH), 2.56 - 2.35 (m, 1H), 2.14 (dd, ./ = 16.1, 5.9 Hz, 1H), 1.96 - 1.74 (m, 3H), 1.66 - 1.37 (m, 3H), 1.28 (d, J= 4.4 Hz, 2H), 1.26 - 1.19 (m, 2H). WF- E (376.1 MHz, CDC13) δ -76.41 (s, 3F, -111.79 (m, 2F). LCMS- t (m/z): [Μ-ίΉ]+ calculated for C24H23F2N O5: 472.17; found: 472.1.
Exam le 30
Preparation of Compound 30
(2S,5R, 13aS)-N-(l -(2,4-difiuorophenyl)cyclopropyl)-8-hydroxy-7,9-dioxo-
2, 3,4,5,7,9 J.3,13a-octahydro-2,5-methanopyri.do[.,2':4,5]pyrazmo[2,l- b] [ 1 ,3]oxazepme- 10-carboxamide
Figure imgf000079_0001
Figure imgf000080_0001
Step 1 and 2
Compound 27-B (150 mg, 0.322 mmol) was dissolved in aeetonitrile (2 mL), AcOH (0.2 mL), and methanesulfonic acid (0.007 mL, 0.108 mmol) at room temperature and the resulting solution was stirred at 65 °C for 20 hours. After the resulting solution was cooled to room temperature, the aminoalcohol 30- A (72.1 mg, chiral, 0.713 mmol), 2C03 (89.4 mg, 0.647 mmol), and aeetonitrile (2 mL) were added to the solution. The resulting mixture was stirred at 65 "C bath for 0.5 hour. After the reaction mixture was cooled to room temperature, it was acidified with 1 N HC1 (~3 mL), diluted with water (-12 mL), and extracted with CH2C12 (x 3). Combined extracts were dried (Na2S04), concentrated, and purified by CombiFlash to obtain 128 mg (84%) of compound 30-B. !H- M (400 MHz, CVK ) δ 10.52 (s, 1H), 8.24 (s, i l l ), 7.61 (td, J = 8.6, 6.6 Hz, 1H), 6.85 - 6.65 (m, 2H), 5.33 (t, J = 4.1 Hz, 1H), 5.25 (dd, J = 9.5, 3.9 Hz, I H), 4.61 (d, J = 3.4 Hz, HI), 4.18 - 4.08 (m, I I I }. 4.02 (s, 3H), 3.99 - 3.87 (m, 1H), 2.12 - 1.91 (m, 4H), 1.85 - 1.69 (m, H I ). 1.55 (ddd, J = 12.3, 4.1 , 2.8 Hz, I H), 1.31 - 1 .14 (m, 4H). 19F-NMR (376.1 MHz, CDCL) δ - 1 1 1.79 (q, J = 8.8 Hz, IF), - 1 12.05 (p, J = 7.9 Hz, IF). LCMS-ESI4 (wi/z): [M+H] + calculated for C24H24F2N3O5: 472.17; found: 472.2. Step 3
A mixture of compound 30-B (128 mg, 0.272 mmoi) and MgBr2 (130 mg, 0.706 mmoi) in MeCN (5 mL) was stirred at 50 °C for 30 minutes and cooled to 0 °C before treating with 1 N HCl (4 mL), After the mixture was diluted with water, the product was extracted with CH2CI2 (x 3), and the combined extracts were dried (Na2S04) and concentrated. The product was purified by CombiFlash to obtain product 30. 1H- MR (400 MHz, CDC1 ) δ 12.27 (s, 1H), 10.52 (s, 1H), 8.16 (s, 1H), 7.61 (td, ./' 8.6, 6.6 Hz, I H), 6.96 - 6.54 (rn. 2H), 5.36 - 5.23 (m, 2H), 4.66 (t, J = 3.1 Hz, 1 H), 4.18 - 4.06 (m, 1H), 3.94 (dd. J 12.8, 9.4 Hz, I H), 2.20 - 1.95 (m, 4H), 1.89 (td, J = 1 1.4, 9.8, 6.7 Hz, I H), 1.70 - 1.54 (m, IH), 1.32 - 1.15 (m, 4H). i9F-NMR (376.1 MHz, CDCI3) δ -1 1 1.87 (q, J - 8.9 Hz, I F), -1 12.21 (p, J = 7.9 Hz, IF). LCMS-ESI4 (m/z): [M+H]+ calculated for C23H22F2N O5: 458.15; found: 458.2.
Example 31
Preparation of Compound 31
(2R,5S)- -(l-(2,4-difiuorophenyl)cyciopropyl)-8-hydroxy-7,9-dioxo- 2,3,4,5,7,9,13, 13 a-octahy dro-2 , 5 -methanopyrido [ Γ ,2 ' : 4 , 5 jpyrazino [2, 1- b] [ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000081_0001
Step 1 and 2
Compound 31-B (123 mg, 81%) was prepared from compound 27-B (150 mg, 0.322 mmol) and the aminoalcohol 31-A (70,3 mg, 0.695 mmol) in a manner similar to that described in step 1 and 2 of the synthesis of compound 30~B. 'H-NM (400 MHz, CDCI3) δ 10.52 (s, 1H), 8.24 (s, 1H), 7.62 (td, 8.6, 6.6 Hz, ill).6.91 - 6.63 (m, 2H), 5.33 (t, J - 4.1 Hz, 1H), 5.25 (dd, J = 9.5, 3.9 Hz, 1H), 4.61 id. J 3.4 Hz, 1H), 4.14 - 4.07 (m, 1H), 4.03 (s, 3H), 3.93 (dd, J= 12.7, 9.5 Hz, 1H), 2.12 - 1.91 (m, 4H), 1.85 - 1.69 (m, 1H), 1.55 (ddd, J = 12.3, 4.1, 2.8 Hz, 1H), 1.31 - 1.14 (m, 4H). "F-NM (376.1 MHz, (IX h) δ -111.79 (q, J = 9.2, 8.7 Hz, IF), -112.03 (h, J= 8.1, 7.5 Hz, IF). LCMS-ES (ni/z): [M+H]+ calculated for C24H24F2N3O5: 472.17; found: 472.1.
Step 3
Compound 31 was prepared from compound 31-B in a manner similar to that described in step 3 of the synthesis of compound 30. 'H- MR (400 MHz, CDCI3) δ 12.26 (s, 1H), 10.49 (s, 1H), 8.13 (s, 1H), 7.58 (td, J = 8.6, 6.5 Hz, 1H), 6.90 - 6.56 (m, 2H), 5.32 (dd, J= 9.4, 4.1 Hz, 1H), 5.27 - 5.22 (m, 1H), 4.64 (t, J= 3.1 Hz, 1H), 4.11 (dd,J= 12.8, 4.0 Hz, 1H), 4.01 - 3.79 (m, 1H), 2.28 - 1.95 (m, 4H), 1.95 - 1.80 (m, 1H), 1.71 (m, 1H), 1.56 (m, 1H), 1.42 - 1.08 (m, 4H). 1 F~NMR (376.1 MHz, CDC1) δ -111.95 (q, J= 8.9 Hz, IF), -112.22 (p, J= 7.9 Hz, IF). LCMS~ESI+ (rn/z): [M+H]÷ calculated for C23H22F2N3O5: 458.15; found: 458.1. le 32
Preparation of Compound 32
(2S,5R)-N-( 1 -(2,4-difluoropheiiyl)cyclobutyi)-8-iiydroxy-7,9-dioxo-2,3 , ,5 ,7,9, 13,13a- octahydro-2,5-methanopyrido[l\2 ,5]pyrazmo[2,l-b][l,3]oxazepine-10-carboxarnide
Figure imgf000082_0001
32
Figure imgf000083_0001
Figure imgf000083_0002
32
A solution of compound 32-A (22.2 mg, 0.069 mmol), compound 3'2-B ( 18.7 mg, 0.102 mmol), and HATU (43 mg, 0.1 13 mmol) in CH2C12 (2 mL) was stirred at room temperature as N,N-diisopropy!ethylamine (DIPEA) (0.075 mL, 0.431 mmol) was added. After 30 minutes, the reaction mixture was diluted with ethyl acetate and washed with water (x 2). After the aqueous fractions were extracted with EA (x 1), the organic fractions were combined, dried, concentrated, and dried in vacuum.
A mixture of the above crude product and MgBr2 (35 mg, 0.190 mmol) o
in eCN (2 mL) was stirred at 50 C bath for 1 hour and cooled to 0 C before being treated with 1 N HQ (- 1 mL.). The resulting solution was diluted with water, and extracted with CH2C12 (x 3). The combined extracts were dried (Na2SC>4), and concentrated. The product was purified by preparative HPLC and freeze-dried to obtain compound 32. 1H- M (400 MHz, CDC13) 8 10.87 (s, IH), -9.3 (br, 1H), 8.35 (s, 1H), 7.50 (id. ./ 8.7, 6.3 Hz, I H), 6.89 - 6.78 (m, I H), 6.72 ( ddd. J 1 1.2, 8.9, 2.6 Hz, I H), 5.48 - 5.12 (m, 2H), 4.72 - 4.60 (m, I H), 4.22 (dd, J ------ 13.0, 4.1 Hz, I H), 3.98 (dd, J =
12.9, 9.4 Hz, I H), 2.68 (m, 4H), 2.33 - 1.98 (m, 6H), 1 .90 (m, 2H), 1.60 (ddd, J = 12.4, 4.1 , 2.7 Hz, IH). "F-NMR (376.1 MHz, CD3CN) 6 -76.39 (s, 3F), -1 10.50 (q, J --- 9,2 Hz, IF), -1 12.65 (p, J = 7.8 Hz, IF). LCMS-ESI+ (m/z): [M+H] calculated for { . ;! ] 1·' · :{ : 472.17; found: 472.0.
Example 33
Preparation of Compound 33 (2S,5R.)-TSi-(l-(254-difiuorophenyl)cyciopentyl)-8-h.ydroxy-7,9-dioxo- 2,3,4,5 J,94343a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l - b] [ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000084_0001
Compound 33 was obtained from compound 32~A and compound 33-A as described in the synthesis of compound 32. 1H- R (400 MHz, CDCI3) 5 10.70 (s, 1H), -9.5 (br, 1H), 8.41 (s, 1H), 7.43 (td, J = 8.9, 6.4 Hz, 1H), 6.85 - 6.76 (m, 1H), 6.72 (ddd, J = 1 1.5, 8.8, 2.6 Hz, 1H), 5.48 - 5.18 (m, 2H), 4.68 (t, J = 3.2 Hz, 1 H), 4.26 (dd, J 13.0, 4.1 Hz, 1H), 4.00 (dd, ./ 13.0, 9.4 Hz, 1H), 2.72 - 2.45 (m, 2H), 2.22 - 1.96 (m, 6H), 1.96 - 1.75 (m, 5H), 1.60 (ddd, J = 12.5, 4.1 , 2.7 Hz, 1H). 19F-NMR (376.1 MHz, CD3CN) 5 -76.41 is. 3F), -107.86 (q, J = 9.4 Hz, I F), -1 13.13 ( p. J = 8.0 Hz, I F). LC S-ES (m/z): | VI · ! ! | calculated for ( i.-(,F N X),: 486.18; found: 485.9. Example 34
Preparation of Compound 34
(2S,5R)-N-(l 2,4-difluorophenyl)cyclohexyl)-8½droxy-7,9-dioxo-2,3,4,5,7,9,13, 13a- octahydro-2,5-methanopyrido[ 1 ',2':4,5]pyrazmo[2, 1 -b][ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000084_0002
Figure imgf000085_0001
Compound 34 was obtained from compound 32~A and compound 34-A as described in the synthesis of compound 32. iH~NMR (400 MHz, CDC13) δ 10.83 (s, 1H), -9.6 (br, ill), 8.44 (s, !!!}.7.37 (td, J= 9.0, 6.4 Hz, 1H), 6.97 - 6.76 (m, 1H), 6.69 (ddd, J = 11.9, 8.8, 2.7 Hz, 1 H), 5.48 -5.18 (m, 2H), 4.68 ft, J = 3.0 Hz, I H), 4.28 (dd, J= 13.1, 4.1 Hz, 1H), 4.03 idd.J 13.0, 9.4 Hz, 1H), 2.60 (d. 13.1 Hz, 2H), 2.29 - 1.96 (m, 4H), 1.95 - 1.77 (m, 4H), 1.77 - 1.65 fm, 4H), 1.61 (ddd, J = 12.5, 4.1, 2.7 Hz, 1H), 1.30 (br, lH). 19F-NMR (376.1 MHz, CD3CN) 6 -76.41 (s, 3F), -107.86 (q../ 9.4 Hz, IF), -113.13 (p, J = 8.0 Hz, IF). LCMS-ESf (m/z): [M+H]+ calculated for ί .,,ΙΚΓ :X. 500.20; found: 500.0.
Exam le 35
Preparation of Compound 35
(2S,5R)-N-(4-(2,4-difluorophenyl)tetrahydro-2H-pyran-4-yl)-8-hydroxy-7,9-dioxo- 2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[l 2':4,5]pyrazino[2,l- b] [ 1 ,3]oxazepiiie- 10-carboxamide
Figure imgf000085_0002
Compound 35 was obtained from compound 32-A and compound 35- A as described in the synthesis of compound 32. ^!-N R (400 MHz, CDCi3) δ 10.95 (s, IH), 8.33 (s, ill). '-7.6 (br, 111).7.38 (td, J 9.0, 6.3 Hz, IH), 6.85 (id../ 8.4, 2.6 Hz, IH), 6.73 (ddd, J = 11.7, 8.6, 2.6 Hz, IH), 5.32 (dt, J = 14.4, 4.0 Hz, 2H), 4.68 (t, J 3.1 Hz, IH), 4.24 (dd, J = 13.0, 3.9 Hz, IH), 4.11 - 3.81 (m, 5H), 2.60 (d, J= 13.7 Hz, 2H), 2.33 - 2.17 (m, 2H), 2.18 - 1.97 (m, 4H), 1.87 (m, IH), 1.61 (dt, J= 12.5, 3.3 Hz, IH). 19F-NMR (376.1 MHz, CD3CN) δ -76.40 (s, 3F), -108.78 (q, J= 10.3, 9.8 Hz, IF), -112.63 (p, J - 8.0 Hz, IF). LCMS-ESI+ (w/z): jVMI; calculated for C25H26F2N3O6: 502.18; found: 502.0.
Example 36
Preparation of Compound 36
(2S,5R)-N-((S)-l-(2,4-difiuorophenyl)-2,2,2-trifluoroethyl)-8-hydroxy-7,9-dioxo- 2,3,4,5,7,9,13,13a-octaliydro-2,5-metlianopyrido[1^2':4,5]pyrazino[2,l- b] [ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000086_0001
32-A 36-A 36
Compound 36 was obtained from compound 32-A and compound 36-A as described in the synthesis of compound 32. 1H-NMR (400 MHz, CDCI3) δ 11.31 (d, J = 9.4 Hz, IH), 8.41 (s, IH), 7.65 - 7.44 (m, IH), 6.95 (ddd, J === 9.6, 5.6, 2.0 Hz, IH), 6.92 - 6.79 (m, IH), 6.15 (h, J = 7.4 Hz, IH), ~6 (br, IH), 5.41 (dd, J= 9.5, 4.0 Hz, IH), 5.31 (t, J= 4.0 Hz, IH), 4.70 (s, IH), 4.34 (dd, J= 12.8, 3.9 Hz, IH), 4.05 (dd, J = 12.9, 9.4 Hz, IH), 2.26 - 1.99 (m, 4H), 1.99 - 1.87 (m, IH), 1.62 (dt, J= 12.6, 3.4 Hz, 1 H). F-NMR (376.1 MHz, CDC13) δ -75.23 (t, J = 6.9 Hz, 3F), -76.33 (s, 3F), - 108.31 (m, IF), -1 12.30 (p, J = 8.0 Hz, I F). LCMS-ES1+ (m/z): [M+H] + calculated for C ,H i J-% :{ : 500.12; found: 500.1.
Example 37
Preparation of Compound 37
(3S,l laR)-N-(l-(2,4-difluorophenyl)cyclopropyl)-6-hydroxy-3-methyl-5,7-dioxo- 2,3,5,7,1 1 ,1 1 a-hexahy droox azolo [3 , 2 -ajpyrido [ 1 , 2 ~d ] pyrazine- 8 -carboxarai de
Figure imgf000087_0001
Figure imgf000087_0002
37-A
Step 1
Methyl 5-(l-(2,4-difi'uorophenyl)cyclopropylcarbamoyl)-l -(2,2- dimethoxyethyl)-3-methoxy-4-oxo-l ,4-dihydropyridine-2-carboxylate (27-B, 0.150 g, 0.32 mmol ) in acetonitrile ( 1.5 mL) and acetic acid (0.2 mL) was treated with methanesuifonic acid (0.05 mL), sealed with a yellow cap, and heated to 70 °C. After 16 hours, the mixture was cooled to afford a crude solution of methyl 5-( l-(2,4- difluorophenyl)cyclopropylcarbamoyl)-l-(2,2-dihydroxyethyl)-3-methoxy-4-oxo-l ,4- dihydropyridine-2-carboxyiate 27-C. LCMS-ESF (m/z): [M+H] calculated for
Figure imgf000088_0001
439; found: 439.
Steps 2 and 3
Methyl 5-(l -(2,4-difluorophenyl)cyclopropylcarbamoyl)-l-(2,2- dihydroxyethyl)-3-methoxy-4-oxo- 1 ,4-dihydropyridiiie-2-carboxylate (27-C, 0.32 mmol, the crude mixture from the previous step) was dissolved in acetoiiitrile (1.5 mL) and acetic acid (0.2 mL). (S)-2-aminopropan-l.-ol (0.048 g, 0.64 mmol) and .2C03 (0.088 g, 0.64 mmol) were added to the reaction mixture. The reaction mixture was sealed and heated to 70 °C. After 3 hours, the reaction mixture was cooled and magnesium bromide (0.081 g, 0,44 mmol) was added. The mixture was resealed and heated to 50 °C. After 10 minutes, the reaction mixture was cooled to 0 °C and 1 N hydrochloric acid (0.5 mL) was added in. Then the reaction mixture was diluted with MeOH (2 mL). After filtration, the crude was purified by Prep-HPLC (30-70% acetoiiitrile I water, 0.1% TFA) to afford Compound 37 as a TFA salt. ¾-NM (400 MHz, Methanol-^) δ 8.31 (s, 1 H), 7.62 (id, J ------ 9.2, 8.7, 6.5 Hz, 1 H), 7.02 - 6.78 (m,
2H), 5.53 - 5.20 (m, 1 H), 4.68 (dd, J ------- 12.3, 4.2 Hz, 1 H), 4.40 (dq, J -------- 19.1 , 6.7 Hz,
2H), 3.98 (dd, J = 12.2, 10.0 Hz, ! H), 3.71 (dd, J ------ 8.3, 6.3 Hz, 1 H), 1 .41 (d, J ------ 6.1
Hz, 3H), 1.22 (s, 4H). 19F-NM (376 MHz, Methanol ~i/4) δ -1 13.66 - -1 13.95 (m, I F), - 1 13.94 - -1 14.29 (m, I F). LCMS-ESF (m/z): [M+H calculated for C21H2oF2 305: 432.; found: 432.
Example 38
Preparation of Compound 38
(18,41 , 12aR;)-N~(2,4~difluoro^^
octahydro-l ,4-methanodipyrido[l ,2-a:r,2'-d]pyrazme-9-carboxamide
Figure imgf000088_0002
38
Figure imgf000089_0001
Step 1
A solution of compound 38-A (1562 mg, 5.799 mmol) (see Example 41b in WO 97/05 139) in THF (10 mL) was stirred at -78 °C as 2.0 M LiBH4 in THF (3.2 mL) was added and the resulting mixture was stirred at room temperature. After 3 hours, additional 2.0 M LiBH4 in THF (3.2 mL) was added and the solution was stirred at room temperature for 17.5 hours. After the reaction mixture was diluted with ethyl acetate and added water slowly, two phases were separated, and the separated aqueous fraction was extracted with ethyl acetate (x 1 ). Two organic fractions were washed with water (x 1), combined, dried (Na2SC>4), and concentrated. The residue was purified by CombiFlash (40 g column) using hexanes - ethyl acetate as eluents to afford compound 38-B. 1H- MR (400 MHz, Chloroform-d) 8 4.1 1 (s, 1H), 3.65 - 3.52 (m, 2H), 3.45 (m, 1H), 2.32 (d, J = 4.1 Hz, 1H), 2.20 (s, 1H), 1.75 - 1.64 (m, 2H), 1.61 (m, 21 1 ). 1.49 - 1.41 (m, 1H), 1.47 (s, 9H), 1 .28 - 1.23 (d, J = 10 Hz, 1 H). LCMS-ESI* (m/z): [M+H]+ calculated for C12H22N03: 228.16; found: 227.7.
Step 2
A solution of compound 38-B (589 mg, 2.591 mmol) and NEt (0.47 mL, 3.369 mmol) in CH2C12 (6 mL) was stirred at 0 °C as MsCl (0.22 mL, 2.842 mmol) was added. After 1 hour at room temperature, the mixture was diluted with ethyl acetate and washed with water (x 2). The aqueous fractions were extracted with ethyl acetate (x
1 ) , and the organic fractions were combined, dried (Na2S04), and concentrated. The residue was purified by Combi Flash (40 g column) using hexanes - ethyl acetate as e!uents to afford compound 38-C. ^l- MR (400 MHz, Chloroform-* ) δ 4.39 - 4.28 (m, 1H), 4.16 (s, 0.4! I k 4.06 (s, 0.6H), 3.98 (dd, J = 10.0, 8.7 Hz, 0.6H), 3.86 (t, J = 9.6 Hz, 0.4H), 3.51 (dd, J = 9.3, 3.7 Hz, 0.6H), 3.43 (dd, J = 9.3, 3.6 Hz, 0.4H), 3.02 (s, 3H), 2.59 (m, 1H), 1.82 - 1.58 (m, 4H), 1.51 - 1.44 (m, 9H), 1.41 (d, J = 14.8 Hz, 1H), 1 .3 1 is. 0.6! I ). 1.29 (s, 0.4H).
Step 3
To a solution of compound 38-C (769 mg, 2.518 mmol,) in DMF (5 mL) was added sodium azide (819 mg, 12.6 mmol). The reaction mixture was stirred at 50 °C for 15 hours, at 80 °C for 5 hours, and at 100 °C for 19 hours. The reaction mixture was diluted with 5% LiCl solution and the product was extracted with ethyl acetate (x
2) . After the organic fractions were washed with water (x 1), the two organic fractions were combined, dried (Na2S04), and concentrated . The residue was purified by
CombiFlash (40 g column) using hexanes - ethyl acetate as eluents to afford compound 38-D. 'H- MM (400 M Hz, Chloroform-*/) δ 4.16 (s, 0.4! I k 4.06 (s, 0.6H), 3.61 (dd, J ------ 12.2, 3.6 Hz, 0.6! ! i. 3.51 (dd, J = 12, 1 , 3.2 Hz, 0.4! I ). 3.38 (dd, J = 9.4, 3.4 Hz,
0.6H), 3.26 (dd, J = 9.8, 3.3 Hz, 0.4 ! I ). 3.06 (dd, J = 12.2, 9.4 Hz, 0.6H), 3.01 - 2.92 (m, 0.4! f k 2.48 (d, J = 5.2 Hz, 1H), 1.82 - 1 .57 (m, 4H), 1 .46 (d, J = 3.0 Hz, 9H), 1.42 (m, IH), 1.28 (m, 0.6H), 1.27 - 1.23 (m, 0.4H).
Step 4
To a solution of compound 38-D (507 mg, 2,009 mmol ,) in ethyl acetate ( 10 mL) and EtOH (10 mL) was added 10% Pd/C (52 mg ). The reaction mixture was stirred under H2 atmosphere for 1.5 hours. The mixture was filtered through celite and the filtrate was concentrated to afford crude compound 38-E. LCMS-ESI+ (m/z): [M+H] ~ calculated for C^H^Chi 227.18; found: 226.8. Step 5
The mixture of crude compound 38-E (206 mg, 0.910 mmol), compound 38-F (330 mg, 0.953 mmol), and 'NaHC03 (154 mg, 1 .833 mmol ) in water (3 mL) and EtOii (3 mL) was stirred at room temperature for 20 hours. After the reaction mixture was diluted with water and extracted with ethyl acetate (x 2), the extracts were washed with water (x 1), combined, dried (Na2S04), and concentrated to afford the crude pyridine product.
The crude residue (388 mg) was dissolved in CH2C1? (4 mL) and 4 N HQ in dioxane (4 mL). After 1.5 hours, additional 4 N HQ in dioxane (4 mL) was added and stirred for 1 hour at room temperature. The mixture was concentrated to dryness, coevaporated with toluene (x 1 ) and dried in vacuum for 30 minutes.
The crude residue and l ,8-diazabicyeloundec-7-ene (DBU) (1.06 mL, 7.088 mmol) in toluene (10 mL) was stirred at 1 10 °C bath. After 30 minutes, the mixture was concentrated and the residue was purified by CombiFlash (40 g column) using ethyl acetate - 20% MeOH/ethyl acetate as eluents to obtain compound 38-G. ' l l-N VI R (400 MHz, Chloroform-d) δ 8.03 (s, I I I . 7.68 - 7.58 (m, 2H), 7.36 - 7.27 (m, 3H), 5.53 (d, J - 9.9 Hz, 1 H), 5.1 1 (d, J = 9.9 Hz, IH), 4.93 (s, H i ). 4.43 - 4.30 (m, 2H), 3.89 (dd, J = 12.2, 3.3 Hz, 1 1 1 . 3.73 (t, J - 12.0 Hz, IH), 3.59 (dd, J = 1 1.9, 3.3 Hz, 1 H), 2.53 (d, J = 2,8 Hz, 1 1 1 . 1 .87 - 1.67 (rn. 4H), 1.55 (d, J = 10.0 Hz, I H), 1 .51 - 1,45 (m, I H), 1.38 (t, J = 7.1 Hz, 3H). LCMS-ES (m/z): | V3 · ! 1 1 calculated for C > ;M .«VO--: 409.18; found: 409.2.
Step 6
The mixture of compound 38~G (232 mg, 0.568 mmol) in THF (3 ml.) and MeOH (3 mL) was stirred at room temperature as 1 N OH (3 mL) was added. After 1 hour, the reaction mixture was neutralized with 1 N HQ (-3.1 mL), concentrated, and the residue was concentrated with toluene (x 3). After the residue was dried in vacuum for 30 minutes, a suspension of the crude residue, 2,4- difluorobenzylamine (86 mg, 0.601 mmol), and HATU (266 mg, 0.700 mmol) were in CH2CI2 (4 mL) and DMF (4 mL) was stirred at 0 °C as N,N-diisopropylethylamine (D1PEA) (0.7 mL, 4.019 mmol) was added. After 45 minutes, additional 2,4- dif3.uorobenzyl.amme (86 mg, 0.559 mmol), HATU (266 mg, 0.700 mmol), and N,N- diisopropylethylarnme (DIPEA) (0.7 mL, 4.019 mmol) were added at room temperature. After 1 .25 hours, the mixture was concentrated to remove most of C! I ··( *! ··, diluted with ethyl acetate, and washed with 5% LiCl (x 2). After the aqueous fractions were extracted with ethyl acetate (x 1 ), the organic fractions were combined, dried (Na2S04), and concentrated. The residue was purified by Comhiflash (40 g column) using ethyl acetate -20%MeOH/ethyl acetate as eluents to afford compound 38-H. ¾~ NMR (400 MHz, Chloroforai-d) δ 10.48 (t, J = 6.0 Hz, 1H), 8.33 (s, IH), 7.62 - 7.51 (m, 2H), 7.40 - 7.27 (m, 4H), 6.87 - 6.75 (m, 2H), 5.39 (d, J = 10.0 Hz, I H), 5.15 (d, J = 10.0 Hz, 1 H), 4.92 (s, 1H), 4.68 - 4.53 (m, 2H), 3.97 (dd, J = 12.5, 3.4 Hz, I I I . 3.77 (t, J = 12.2 Hz, I H), 3.55 (dd, J = 12.1 , 3.3 Hz, I I ! ;·. 2,53 (d, J = 3.1 Hz, 1 H), 1.88 - 1.62 (m, 41 ! }. 1.59 - 1 .42 (m, 2H). 19F- MR (376 MHz, Chlorofom -d) δ -1 1 2. 1 7 (q, J = 7.6 Hz, I F), - 1 14.79 (q, J = 8.6 Hz, I F). LCMS-ESr (m/z) : [M+H]+ calculated for C28H26F2N304: 506.19; found: 506.2.
Step 7
Compound 38~M (240 rag, 0.475 mmol) was dissolved in TFA (3 mL) at room temperature for 30 minutes, and the solution was concentrated. The residue was purified by CombiFlash (40 g column) using CH2Cl2-20% MeOH in { ! l.-i I - as eluents. After the col lected product fractions were concentrated, the residue was triturated in MeCN (~2 mL) at 0 °C for 15 minutes, and the solids were filtered and washed with MeCN. The collected solids were dried in vacuum to afford compound 38.
The filtrate was concentrated, and the residue was dissolved in MeCN (~1 mL) and water (~1 mL) by heating. The solution was slowly cooled to room temperature and then in ice bath for 15 minutes. The solids were filtered and washed with MeCN, and dried in vacuum to afford additional compound 38. IH-NMR (400 MHz, Chloroforn -d) δ 1 1 .68 (s, 1 1 ! }. 10.42 (s, I H), 8.27 (s, i l l ). 7.41 - 7.31 (m, I H), 6.86 - 6.73 (m, 2H), 4.90 (d, J = 2.5 Hz, I H), 4.71 - 4.53 (m, 2H), 4.07 (d, J = 10.6 Hz, IH), 3.90 - 3.67 (m, 2H), 2.68 (s, I H), 2.01 (s, I H), 1 .97 - 1.80 (m, 3H), 1.80 - 1.62 (m, 2H). 19F~NMR (376 MHz, Chlorofomi-d) 8 -1 12.28 (m, IF), 14.74 (m, IF). LCMS- ESI+ (m/z): [M+H calculated for C21H19F2N3O4: 416.14; found: 416.3. Examples 39 and 40
Preparation of Compounds 39 and 40
(2R,3S,5R,13aS)-N-(2,4-difluorobenzyl)-8-hydroxy-3-methyl-7,9-dioxo- 2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[ ,2':4,5]pyrazino[2, 1 - ,3]oxazepine- 10-carboxamide 39 and (2S,3R,5S,13aR)-N-(2,4-difluorobenzyl) hydroxy-3-met yl-7,9-dioxo-2,3,4,5,7,9,13,13a-octahydro-2,5- metlianopyrido[r,2':4,5]pyrazino[2, 1 -b] [1 ,3]oxazepine- 10-carboxamide 40
Figure imgf000093_0001
Step 1
Cuprous cyanide (290 mg, 3,27 mmol) was suspended in 3.3 mL THF and cooled to -78 °C. A 1.6M solution of Me Li (4.1 mL, 6.56 mmol) in diethyl ether was added dropwise, the reaction solution was allowed to warm to room temperature over the course of 2 hours, and recooled to -78 °C. Tert-butyl (lR,3R,5S)-6- oxabicyclo[3.1.0]hexan-3-ylcarbamate (330 mg, 1.66 mmol) was added dropwise in 3.3 mL THF, followed by boron trifluoride diethyl etlierate (0.25 mL, 1.99 mmol), allowed to warm to -30 °C over 30 minutes, and stirred between -35 °C and -25 °C for one hour. The reaction solution was then wanned to room temperature and quenched with a mixture of saturated H3(aq)/ H4(aq), extracted to EtOAc, washed with brine, dried over MgS()4, filtered, concentrated, and purified by SGC (0-10% EtOH/DC ) to afford racemic tert-butyl (l S,3S,4S)-3-hydroxy-4-methylcyclopentylearbamate. 3H- NMR (400 MHz, Chloroform-^ 6 5.16 (s, 1 1 1 ). 3.98 (s, IH), 3.74 (q, ,/ 4.3 Hz, IH), 3.65 (q, J = 7.0 Hz, 1H), 2.23 (dt, J = 14.0, 7.0 Hz, 1H), 1.98 (dt, J= 13.3, 7.0 Hz, 1H), 1.89 - 1.79 (m, IH), 1.58 - 1.44 (m, IH), 1.38 (s, 9H), 1.18 (t, J = 7.0 Hz, 1H), 0.91 (d, ./ = 7.0 Hz, 3H).
Step 2
3 mL HCi/dioxane (4M, 12 mmol ) was added to a solution of racemic tert-butyl (l S,3S,4S)-3-hydroxy-4-methylcyclopentylearbamate (182 mg, 0.85 mmol) in 3 mL dioxane. The reaction mixture was stirred at room temperature for 2 hours, concentrated and twice chased with toluene to afford racemic (lS,2S,4S)-4-ammo-2- methylcyclopentanol.
Step 3
Methyl 5-(2,4-difluorobenzylcarbamoyl)- 1 -(2,2-dihydroxyethyl)-3- methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-C, 310 mg, 0.75 mmol), racemic (lS,2S,4S)-4-amino-2-methylcyclopentanol (115 mg, 0.76 mmol), and potassium carbonate (232 mg, 1 ,68 mmol) were taken up in 3.8 mL. acetonitrile/0,2 mL acetic acid and stirred at 90 °C for 2 hours, after which the reaction mixture was partitioned between DCM and brine, the aqueous phase extracted to DCM, combined organic phases dried over MgS04, filtered, concentrated, and purified by SGC (0-10% EtOH/DCM) to afford intermediate 39-A.
Step 4
Intermediate 39-A (190 mg) was separated by chiral Prep-HP LC on a Lux Cellulose-2 column using 9: 1 ACN:MeOH as eluent to afford Intermediates 39-B (first eluting peak) and 40-A (second during peak) in enantioenriched form. For intermediate 39-B: (absolute stereochemistry confinned by XRay crystallography), Chiral HPLC retention time ::: 3.98 minutes (Lux Cellulose-2 IC, 150 x 4.6 mm, 2 mL/min 9: 1 ACN:MeOH). For intermediate 40-A: (absolute stereochemistry confirmed by XRay crystallography), Chiral HPLC retention time = 6.35 minutes (Lux Cellulose- 2 IC, 150 x 4.6 mm, 2 mL/min 9: 1 ACN:MeOH).
Step 5 a
Magnesium bromide (68 mg, 0.37 mmol) was added to a solution of intermediate 39-B (83 mg, 0.18 mmol) in 2 ml. acetonitrile. The reaction mixture was stirred at 50 °C for 1 hour, acidified with 10% aqueous HQ, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted to dichloromethane. The combined organic phases were dried over gS04, filtered, concentrated, and purified by silica gel chromatography (0-10% EtOH/DCM) to afford compound 39. XH- NMR (400 MHz, Chloroform-d) δ 12.32 (s, lH), 10.36 (s, 1H), 8.29 (s, 1 H), 7.44 - 7.33 (m, 1H), 6.88 - 6.76 (m, 21 1 ). 5.37 (dd, J = 9.5, 4.1 Hz, 1H), 5.28 (t, J = 5.3 Hz, 1H), 4.63 (d, J = 5.9 Hz, 2H), 4.23 (d, J - 23.0 Hz, 2H), 3.99 (dd, J = 12.7, 9.5 Hz, I F), 3.72 (q, J - 7.0 Hz, 1H), 2.51 (dq, J = 13.7, 6.8, 6.1 Hz, 1H), 2.15 (ddd, J = 14.7, 8.3, 2.3 Hz, 1 H), 1.94 i d. J = 12.7 Hz, 1 H), 1.77 (ddd, .1 - 12.7, 4.0, 2.9 Hz, 1 H), 1 .61 (dt, J = 14.6, 5.2 Hz, 2H), 1.24 (t, J = 7.0 Hz, 1 H), 1.09 (d, J = 7.2 Hz, 3H). LC .VIS-ESI (m/z): [ VI H i calculated for C · ,! 1Λ Λ :Ch: 446.15; found: 446.2.
Step 5b
Magnesium bromide (59 mg, 0.32 mmol) was added to a solution of intermediate 40-A (70 mg, 0.15 mmol) in 2 ml. acetonitrile. The reaction mixture was stirred at 50 °C for 1 hour, acidified with 10% aqueous HQ, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted to dichlorometh ane. The combined organic phases were dried over MgS0, filtered, concentrated, and purified by silica gel chromatography (0-10% EtO /DCM) to afford compound 40.3"H- NMR (400 MHz, Chloroform-i) δ 12.32 (s, 1H), 10.36 (s, Hi).8.29 (s, III}.7.44 - 7.33 (m, 1H), 6.88 - 6.76 (m, 2H), 5.37 (dd, J= 9.5, 4.1 Hz, 1H), 5.28 (t, J = 5.3 Hz, 1H), 4.63 (d, J= 5.9 Hz, 2H), 4.23 (d, J= 23.0 Hz, 2H), 3.99 (dd.J 12.7, 9.5 Hz, 1H), 3.72 (q, J= 7.0 Hz, 1H), 2.51 (dq, J= 13.7, 6.8, 6.1 Hz, 1H), 2.15 (ddd, J= 14.7, 8.3, 2.3 Hz, 1H), 1.94 (d, J ------ 12.7 Hz, 1H), 1.77 (ddd, J = 12.7, 4.0, 2.9 Hz, 1H), 1.61 (dt, J
- 14.6, 5.2 Hz, 2H), 1.24 (t, J ------- 7.0 Hz, 1H), 1.09 id../ 7.2 Hz, 3H). LCMS-ESI+ (m/z): \\\ l!j calculated for C-l Ι,,ί' :(),: 446.15; found: 446.2.
Preparation of Compound 41
(lR,4S 2aR)-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)-l ,2,3,4,6,8,12,12a- octahydro-l,4-methanodipyrido[l,2-a:r,2'~d]pyraziiie-9-carboxamide
Figure imgf000096_0001
41
Figure imgf000097_0001
Figure imgf000097_0002
Steni
A solution of the 41-A (2020 mg, 7.463 mmol) (prepared by the same method as 38-A) in THF (14 mL) was stirred at 0 °C as 2.0 M LiBH4 in THF (7.5 mL, 15 mmol) was added. After the resulting mixture was stirred at rt for 21 h, it was cooled at 0 °C and diluted with EA. before water was added slowly to quench. After two phases were separated, the aqueous fraction was extracted with EA (x 1) and the two organic fractions were washed with water (x 1 ), combined, dried (Na2S04), and concentrated. The residue was purified by CombiFlash (120 g column) using hexanes - EA as eluents to get 41-B. LCMS-ESI* (m/z): [M-C4¾+H]+ calculated for C8Hi4N03: 172.10; found: 171.95.
Step 2
A 100-mL round bottom flask was charged with reactant 41 -B (1.6 g, 7.05 mmol) and triethylamine (0.94 g, 9.3 mmol) in DCM (20 mL). ethanesulfonyl chloride (0.91 g, 8,0 mmol) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for 3 hours. The mixture was diluted with EA (100 mL) and washed with water (2x). The aqueous fractions were extracted with EA (lx), and the organic fractions were combined, dried (Na2S04), and concentrated. The residue was purified by Combi Flash (120 g column, cartridge used) using hexanes - EA as eluents to afford 41-C. LCMS-ESI (m/z): [M+Hf calculated for CJ 8H39F2N207: 306; found: 306,
Step 3
A 100-mL round bottom flask was charged with reactant 41-C (2.1 g, 6.9 mmol) and sodium azide (2.3 g, 34.5 mmol) in DMF (10 mL). Then the reaction mixture was stirred at 100 C for overnight. The mixture was diluted with EA (100 mL) and washed with water (2x). The aqueous fractions were extracted with EA (lx), and the organic fractions were combined, dried (Na2S04), and concentrated. The residue was purified by Combi Flash (120 g column, cartridge used) using hexanes - EA as eluents to afford 41-D. L€MS~ESI+ (m/z): [M+H]+ calculated for C18Hi9F2N207: 253; found : 253.
Step 4
To a solution (purged with N2) of reactant 41-D (1 .3 g) in EA (20 mL) and EtOH (20 mL) was added Pd/C (130 mg). The mixture was stirred under H? for 3 hours. The mixture was filtered through celite and the filtrate was concentrated to afford compound 41-E. L-CMS-ESI (m/z): [M+H]+ calculated for Cj 8FI j9F2N2Q7: 227; found : 227.
Step 5
A 100-mL round bottom flask was charged with reactant 41-E (1.05 g, 4.62 mmol ) and reactant 38-F (1.6 g, 4.62 mmol) in Ethanol (20 mL). Sodium bicarbonate (0.77 g, 9.2 mmol) in water (20 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature overnight. The mixture was diluted with EA (100 ml.) and washed with water (2xj. The aqueous fractions were extracted with EA (l x), and the organic fractions were combined, dried (Na2S04), and concentrated. The crude product (2.4 g) was used for next step without further purification. LCMS-ESI+ (m/z) [M+H]+ calculated for C18H19F2N2O7: 556; found: 556.
A 100-mL round bottom flask was charged with the crude product from the previous reaction in 4 N HQ /dioxane (24,7 mL). Then the reaction mixture was stirred at room temperature for 1 hour. After concentration, the intermediate (2.1 g) and DBU (3.27 g, 21.5 mmol) in toluene (30 mL) was heated to 110 °C with stining for 1 hour. After concentration, the residue was purified by CombiFlash (120 g column) using hexanes - ethyl acetate as eluents to afford 41-F. LCMS-ESI+ (m/z): [MrH] calculated for ( i J i ;...i: >N■()· ; 409; found: 409. Step 6
A 100-mL round bottom flask was charged with reactant 41-F (0.5 g, 1.22 mmol) in THF (5 mL) and MeOH (5 mL). 1 N OH (3.7 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was acidified by adding 1 N HQ (3.7 mL), concentrated to remove most of organic solvents, and extracted with EtOAc (2 X). The organic layers were combined, dried iXa ·ΗΟ : }, and concentrated to afford compound 41-G.
Step 7
A 100-mL round bottom flask was charged with reactant 41-G (0.14 g, 0.37 mmol ), (2,4,6-trifluorophenyl)methanamine (0.12 g, 0.73 mmol), N,N- diisopropylethyiamme (DIPEA) (0.24 g, 1.84 mmol) and HATU (0.28 g, 0.74 mmol) were dissolved in DCM (5 mL). The reaction mixture was stirred at room temperature for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated Na C03 (2x), saturated NH4CI (2x) and dried over Na2S04. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford compound 41-H. LCMS-ESf (m/z): [M+H]+ calculated for ( i J I ,oF A .()-: 524.5; found: 524.5.
Step 8
A 50-mL round bottom flask was charged with reactant 41-H (0.13 g, 0.25 mmol) in TEA (2 mL). The reaction mixture was stirred at room temperature for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 41. ^-NMR (400 MHz, Chloroform-d) δ 11.61 (s, IH), 10.70 - 10.01 (m, 1H), 8.26 (s, III}.6.65 (t, J = 8.1 Hz, 2H), 4.88 (s, III}.4.65 (dd, J = 6.1, 2.4 Hz, 2H), 4.07 (d, J - 10.9 Hz, 1H), 3.93 - 3.58 (m, 2H), 2.67 (d, J = 3.1 Hz, IH), 2.08 - 1.41 (m, 7H). 19F-NMR (376 MHz, Chloroform-d) δ 409.22 (d, J = 11.6 Hz, IF), -111.04 - -112.79 (m, 2F). LCMS~E8I+ (m/z): [M+H calculated for CMH 434.; found: 434.
Example 42
Preparation of Compound 42
(2 ,5S,13aI )-8-hydroxy-7,9-dioxo-N-(2,4,6-trifluorohe
octahydro-2,5-methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b][ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000100_0001
42
Figure imgf000101_0001
Step 1
l-(2,2-dimethoxyethyl)-5-methoxy-6-(methoxycarbonyl)-4-oxo-l ,4- dihydropyridine-3-carboxylic acid (3.15 g, 10 mmol) in acetonitrile (36 mL) and acetic acid (4 mL) was treated with methanesuffhnic acid (0.195 mL, 3 mmol) and placed in a 75 deg C bath. The reaction mixture was stirred for 7 h, cooled and stored at -10 °C for 3 days and reheated to 75 °C for an additional 2 h. This material was cooled and carried on crude to the next step.
Step 2
Crude reaction mixture from step 1 (20 mL, 4.9 mmol) was transferred to a flask containing (lR,3S)-3-aminocyclopentanol (0.809 g, 8 mmol). The mixture was diluted with acetonitrile (16.8 mL), treated with potassium carbonate (0.553 g, 4 mmol) and heated to 85 °C. After 2 h, the reaction mixture was cooled to ambient temperature and stirred overnight. 0.2M HQ (50 mL) was added, and the clear yellow solution was extracted with dichloromethane (2x150 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated to 1.49 g of a light orange solid. Recrystallization from dichloimethane:hexanes afforded the desired intermediate 42 A: LC S-ESI (m/z): [M+H]+ calculated for Ci5Hi7N206: 321.1 1 ; found: 321.3. Step 3
Intermediate 42-A (0.225 g, 0.702 mmol) and (2,4,6- trifluorophenyl)methanamine (0.125 g, 0.773 mmol) were suspended in acetonitrile (4 mL) and treated with N,N-diisopropylethylamine (DIPEA) (0.183 mmol, 1.05 mmol). To this suspension was added (dimethyiammo)- V,A/-dimethyi(3H-[l ,2,3]triazolo[4,5- &]pyridm~3-yiox.y)methammimum hexafluorophosphate (HATU, 0.294 g, 0.774 mmol). After 1.5 hours, the crude reaction mixture was taken on to the next step. LfJMS-ESlT (m/z): [M+H calculated for (\ ,l l.,, i \\:0< : 464.14; found: 464.2.
Step 4
To the crude reaction mixture of the previous step was added MgBr2
(0.258 g, 1.40 mmol). The reaction mixture was stirred at 50 °C for 10 minutes, acidified with 10% aqueous HC1, and extract twice with dichloromethane. The combined organic phases were dried over MgS04, filtered, concentrated, and purified by silica gel chromatography (EtOH/dichlormethane) followed by HPLC (ACN H2O with 0.1 % TFA modifier) to afford compound 42: 1H~ M (400 MHz, DMSO-</6) δ 12.43 (s, 1H), 10.34 (t, J = 5.7 Hz, IH), 8.42 (s, 1H), 7.19 (t, J = 8.7 Hz, 2H), 5.43 (dd, ./' 9.5, 4.1 Hz, I H), 5.08 (s, i l l ). 4.66 (dd, ./ 12.9, 4.0 Hz, IH), 4.59 (s, 1 1 1 ). 4.56 4.45 (m, 2H), 4.01 (dd, J = 12.7, 9.7 Hz, IH), 1.93 (s, 4H), 1.83 (d, J ------ 12.0 Hz, I H),
1.56 (dt, J = 12.0, 3.4 Hz, I H). LCMS-ESI+ (m/z): [M+H]+ calculated for { · Ί ί ] ΝΓ :Χϋ : 450.13; found: 450.2.
Example 43
Preparation of Compound 43
( 12aR)-N-((R)- 1 -(2,4-difluorophenyl)ethyl)-7-hydroxy-6,8-dioxo- 1 ,2,3 , ,6,8,12,12a- octahydro-l ,4-methanodipyrido[I ,2-a:r,2'-d]pyrazine-9-carboxarnide
Figure imgf000103_0001
Figure imgf000103_0002
Step 1
A 100-mL round bottom flask was charged with reactant 41~G (0.14 g,
0.37 rnmol), (R.)-l-(2,4-difluorophenyl)ethanamine (0.12 g, 0.74 rnmol), N,N- diisopropylethyl amine (0.24 g, 1 .84 rnmol) and HATU (0.28 g, 0.74 rnmol) and were dissolved in DCM (5 mL). The reaction mixture was stirred at room temperature for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated NaHC03 (2x), saturated NH4C (2x) and dried over Na2S04. After concentration, the cmde was purified by column chromatography on silica gel with hexane-EtOAc to afford compound 43-A. LCMS~ESI+ (m/z): [M+R calculated for
Figure imgf000103_0003
520; found: 520.
Steps 2
A 50-mL round bottom flask was charged with reactant 43-A (0.14 g,
0.27 rnmol) in TFA (2 mL). The reaction mixture was stirred at room temperature for 30 minutes. After concentration, the cmde was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 43. !H- MR (400 MHz, Chiorofomi-d) 8 1 1 .65 (s, ! 1 1 ).. 10.57 (s, 1 1 1 ). 8.22 (s, 1 1 1 ). 7.31 (m, 1 1 1 ). 6.99 - 6.62 (m, 2H), 5.64 - 5.32 (m, I I I . 4.90 (d, J - 2.7 Hz, 1H), 4.04 (d, J = 1 1 .5 Hz, 1H), 3.93 - 3.63 Cm. 2H), 2,67 (s, 1 H), 2,08 - 1 .40 ( . 9H). F-NMR (376 MHz, Chioroform-d) δ - 1 13,09 (m, IF), -1 15.01 (m, IF). LCMS-ES1+ (m/z): j VI H i calculated for C2iH2oF2N305: 430.; found: 430.
Preparation of Compound 44
(13aS)-8-hydroxy-7,9-dioxo-N 23,4-trifluorobenzyl)-2 ,4,5J,9 3J3a-octahydro
2,5-methanopyrido[ 1 \2':4,5]pyrazmo[2, 1 -b] [ 1 ,3]oxazepine-l 0-carboxamide
Figure imgf000104_0001
Step 1
Compound 15-B (40 mg, 0.12 rnmoi) was taken up in 1 mL acetomtrile and treated with 2,3,4-triiluorobenzyiamine (29 mg, 0.18 mmol), HATU (53 mg, 0, 14 mmol), , -diisopropylethylamine (DI PEA) (20 mg, 0.16 mmol), and stirred at room temperature for 2 hours, after which LCMS analysis revealed complete consumption of compound 15-B and formation of intermediate 44-A. The reaction mixture was earned onto the next step. Step 2
To the crude reaction solution of the previous step was added MgBr2 (63 mg, 0.34 mmol). The reaction mixture was stirred at 50 °C for one hour, acidified with 10% aqueous HCL partitioned between the aqueous and dichloromethane, and the aqueous phase extracted to dichloromethane. The combined organic phases were dried over MgSQ4, filtered, concentrated, and purified by HPLC (ACN/H20 with 0.1% TFA modifier) to compound 44. 1H-NM (400 MHz, DMSO-</6) δ 12.45 (s, 1H), 10.38 (t, J - 6.0 Hz, i l l ), 8.43 (s, 1 H), 7.27 (q, J = 9.2 Hz, 1H), 7.16 (q, J = 8.5 Hz, 1 1 1 ). 5.42 (dd, J 9.5, 4.0 Hz, lH), 5.08 (s, 1 H), 4.76 - 4.47 (m, 4H), 4.01 (dd, J ------ 12.8, 9.7 Hz, 1 H), 1.92 (s, 41 I K 1.82 (d, ./ 12.1 Hz, 1 H), 1.55 (dt, J = 12.2, 2.9 Hz, i l l ). LOIS- ES I (m/z): [M+H] calculated for <V , I I T .-V - 450.13; found: 450.2.
Example 45
Preparation of Compound 45
( 13aS)-8-hydroxy-7,9-dioxo-N-(2,4,6-trifluorobenzyl)-2,3 ,4,5 ,7,9,13, 13a-octahydro- 2,5-methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b] [ 1 ,3]oxazepine-l 0-carboxamide
Figure imgf000105_0001
Step 1
Compound 15-B (38 rag, 0, 12 mmol) was taken up in 1 mL acetonitrile and treated with 2,4, 6-trifluorobenzy famine (34 mg, 0.21 mmol), HATU (50 mg, 0.13 mmol), N,N-dii.sopropylethylamine (DIPEA) (23 mg, 0, 1 8 mmol), and stirred at room temperature for 2 hours, after which LCMS analysis revealed complete consumption of compound 15-B and formation of intermediate 45-A. The reaction mixture was carried onto the next step.
Step 2
To the cmde reaction solution of the previous step was added MgBr2 (55 mg, 0.30 mmoi). The reaction mixture was stirred at 50 °C for one hour, acidified with 10% aqueous HC1, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgS04, filtered, concentrated, and purified by HPLC (ACN/¾0 with 0.1% TFA modifier) to afford compound 45. 'H-NM (400 MHz, DMSO-cfe) δ 12.37 (s, 1 H), 10.37 - 10.25 (m, 1H), 8,37 (s, 1 1 1 ), 7.14 (t, J -- 8.7 Hz, 2H), 5.37 (dd, J = 9.5, 4.0 Hz, 1H), 5,02 (s, 1H), 4,66 - 4.40 (m, 4H), 3.95 (dd, J ------ 12.8, 9.6 Hz, 1H), 1 ,87 (s, 4H),
1 .77 (d, ./' 1 1 .9 Hz, 1H), 1.50 (dt, ./ 1 1 .8, 3.2 Hz, 1 H). LCMS-ESf (ni/z): \ \\ i i j calculated for C > ; i ! :" Λ ><);: 450.13; found: 450.2. Example 46
Preparation of Compound 46
(13aS)-N-i2,6-difluorobenzy^
methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b] [1 ,3]oxazepine-l 0-carboxamide
Figure imgf000106_0001
Figure imgf000107_0001
Step 1
Compound 15-B (38 mg, 0.12 mmol) was taken up in 1 mL acetomtrile and treated with 2,6-difluorobenzylamme (19 mg, 0.14 mmol), HATU (56 mg, 0.15 mmol), , -diisopropylethylamine (DI PEA) (20 mg, 0.15 mmol), and stirred at room temperature for 90 minutes, after which LCMS analysis revealed complete consumption of compound A and formation of intermediate 46-A. The reaction mixture was carried onto the next step.
Step 2
To the crude reaction solution of the previous step was added MgBr2 (50 mg, 0.27 mmol). The reaction mixture was stirred at 50 °C for one hour, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgS04, filtered, concentrated, and purified by HPLC (ACN/H JO with 0.1% TFA modifier) to afford compound 46. 'H-NM (400 MHz, DMSG-<i6) δ 12.37 (s, 1H), 10.33 - 10.26 (m, 1H), 8.37 (s, 1H), 7.39 - 7.29 (m, 1H), 7.05 (t, J = 7.9 Hz, 2H), 5.37 (dd, J = 9.5, 4.1 Hz, 1 H), 5.02 (s, 1 H), 4.66 - 4.45 (m, 4H), 3.95 (dd, J = 12.7, 9.6 Hz, 1 H), 1 .87 (s, 4H), 1.77 (d, J -- 12.0 Hz, 1H), 1.50 (dt, J ------ 12.2, 3.5 Hz, 1 H).
LCMS-ESI+ (m/z): [ +H]4 calculated for ί.,,,Γ Λ.,Ο--: 432, 14; found: 432.2, Example 47
Preparation of Compound 47
(l R,4S, 12aR)-N<2,4-difliiorobenzyl)-7-hydroxy-6,8-dioxo-l , 2,3,4,6,8,12, 12a- octahydro-l ,4-methanodipyrido[l ,2-a: ,2'-d]pyrazine-9-carboxamide
Figure imgf000108_0001
47
Step 1
The crude acid 41-G (0.45 g, 1.18 mmol), 2,4-difluoberizylamme (0.35 g, 2.44 mmol), N,N-diisopropylethylamine (DIPEA) (0.79 g, 6.1 1 mmol) and HATU (0.93 g, 2.44 mmol) were dissolved in DCM (10 mLj. The reaction mixture was stirred at room temperature for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated NaHC03 (2x), saturated NEUCf (2x) and dried over Na2S()4. After concentration, the crude was purified by column chromatography on silica gei with hexane-EtOAc to afford compound 47-A. LCMS-ESI+ (m/z): [M+H]+ calculated for Ci J ! ;.)!·' .N >()··: 506; found: 506. Step 2
A 50-mL round bottom flask was charged with reactant 47- A (0.5 g, 0.99 mmol ) in TFA (6 mL). The reaction mixture was stirred at room temperature for 30 mmutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 47. 1H NMR (400 MHz, Chloroform-d) δ 1 1.70 (s, 1H), 10.44 (s, 1H), 8.29 (s, 1H), 7.60 - 7.29 (m, 1H), 6.95 - 6.58 (m, 2H), 4.10 (s, 1H), 4.02 - 3.54 (m, 3H), 2.68 (d, J = 3.1 Hz, 1H), 2.00 - 1.40 (m, 8H). 1 F NM R (376 MHz, Chloroform-d) δ -1 12.31 (d, J = 8.0 Hz, I F), -1 14.77 (d, J = 8.4 Hz, I F). LCMS-ESf (m/z): [ M I I ! calculated for C21H20F2N3O5: 416.; found: 416.
Example 48
Preparation of Compound 48
(lS,4R,12aS)-N-(2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12a- octahydro-l ,4-methanodipyrido[l ,2-a:r,2'-d]pyrazine-9-carboxamide
Figure imgf000109_0001
48-B was prepared analogous!)' to 55-H in Example 55, substituting 48- A for 55-A. Compound 48 was prepared as described for compound 38 in Example 38, substituting 48-B for 38-B to afford compound 48. !H- M (400 MHz, Chloroform- d) 6 1 1.79 (s, 1H), 10.44 (m, 1H), 8.33 (s, 1 H), 7.42 - 7.31 (m, 1 H), 6.86 - 6.74 (m, 2H), 4.74 (s, 1H), 4.63 (d, J = 5.8 Hz, 2H), 4.19 (m, 1H), 4.07 - 4.03 (m, 21 1 ). 2.83 (s, 1H), 1.92 - 1.68 (m, 6H). 19F NMR (376 MHz, Chloroform-d) δ -1 12.3 (m, IF), -1 14.8 (m, IF). LCMS-ESI+ ( n/z): \ \\ \ \ \ calculated for ( \ : H )l: ..VO ,: 416.14.; found: 41 6.07. le y
Preparation of Compound 49
(2S,5R,13aS)-8-hydroxy-7,9-dioxo-N-((3-(trifluoiOmethyl)pyridin-2
2,3,4,5 J,94343a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l - b] [ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000110_0001
49
Figure imgf000110_0002
Compound 15-B (44 mg, 0.14 mmol) was taken up in 1 mL acetonitrile and treated with (3-(trifluorornethyl)pyridin-2-yl)methanamine (38 mg, 0.1 8 mmol, HC1 salt), HATU (69 mg, 0.18 mmol), Ν,Ν-diisopropyiethyiamine (DIPEA) (0.07 mL, 0.40 mmol), and stirred at room temperature for 1 hour, after which LCMS analysis revealed complete consumption of compound 15-B and formation of intermediate 49- A. The reaction mixture was carried onto the next step. Step 2
To the crude reaction solution of the previous step was added MgBr2 (51 mg, 0.28 mmol). The reaction mixture was stirred at 50 °C for 90 minutes, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgS04, filtered, concentrated, and triturated by methanol followed by diethyl ether to afford compound 49. 1H-NMR (400 MHz, DMSO-d6) δ 12.42 (s. 1H), 10.80 - 10.70 (m, 1H), 8.83 (d, J = 5.0 Hz, ! H), 8.44 (s, IH), 8.19 (d, J = 8.6 Hz, ! H), 7.56 (dd, J ------ 7.7, 5.2 Hz, IH), 5.43 (dd, J ------ 9.5, 4.0 Hz, 1 H), 5.08 (s, IH), 4.86 - 4.80 (m, 2H), 4.67 (dd, J = 12.9, 4.0 Hz, IH), 4.59 (s, IH), 4.02 (dd, ./ 12.6, 9.8 Hz, I H), 1.93 (s, 4H), 1.82 (d, J = 12.1 Hz, I H), 1.60 - 1 .52 (m, I H). LCMS-ESi+ (m/z): [M+H]+ calculated for C21H20F3N4O5 : 465.14; found: 465.2.
Examples 50 and 51
Preparation of Compounds 50 and 51
N-(2,4-difluorobenzyl)-9-hydroxy-8,10-dioxo-2,3,5,6,8,10J4,14a^>ctahydro-2,6- methanopyrido[lV2':4,5]pyrazino[2,l-b][l ,6,3]dioxazocine-l .1 -carboxaraide 50 and 51
Figure imgf000111_0001
Figure imgf000112_0001
Methyl 5-(2,4-difluorobenzylcarbamoyl)- 1 -(2,2-dihydroxyethyl)-3- methoxy-4-oxo-l ,4-dihydropyridine-2-carboxyiate (i~C, 392 mg, 0,95 rnmol) (Example 87), racemic cw-5-ammotetrahydro-2H-pyran-3-ol (WO 2012/145569 Bennett, B. L. et al, filed April 20, 2012 ) (1 12 mg, 0.95 rnmol), and potassium carbonate (134 mg, 0.97 mmol) were taken up in 3.8 mL acetonitrile/0.2 mL acetic acid and stirred at 90 °C for 90 minutes, after which the reaction mixture was partitioned between DCM and brine, the aqueous phase extracted with DCM, combined organic phases dried over MgS04, filtered, concentrated, and purified by SGC (0-10% EtOH/DCM) to afford intermediate 50-A.
Intermediate 5Θ-Α (40 mg) was separated by chiral SFC on a Chiralpak IC column using 10% DMF in supercritical carbon dioxide as eluent to afford Intermediates 5Θ-Β (first eluting peak) and 51-A (second eluting peak) in enantioenriched form. For intermediate 50-B: (absolute stereochemistry unknown), Chiral HPLC retention time = 1 1.48 minutes (Chiralpak IC, 150 x 4.6 mm, 1 mL/rnin MeOH), For mtermediate 51-A: (absolute stereochemistry unknown), Chiral HPLC retention time ::: 14.35 minutes (Chiralpak IC, 150 x 4.6 mm, 1 rnL/min MeOH).
Step 3a
Magnesium bromide (12 mg, 0.06 rnmol) was added to a solution of intermediate 50~B (10.5 mg, 0.02 mmol, absolute stereochemistry unknown) in 1 rnL acetonitrile. The reaction mixture was stirred at 50 °C for 1 hour, acidified with 10% aqueous HCl, partitioned between the aqueous and dichlorom ethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgS04, filtered, concentrated, and purified by HPLC1 (ACN/H20 with 0.1 % TFA modifier) to afford compound 50. 1 H-NMR (400 MHz, Chioroforn ) δ 10.47 (t, J 5.H Hz, 1H), 8.42 (s, 1H), 7.35 (q, J = 8.6, 8.2 Hz, 1 H), 6.81 (q, J = 8.7, 8.0 Hz, 2H), 6.41 (dd, J = 10.0, 3.6 Hz, IH), 4.79 (s, IH), 4.65 (s, 2H), 4.36 - 4.26 (m, 21 1 ). 4.20 - 4.08 (m, 2H), 3.98 (dd, J = 12.4, 10.2 Hz, IH), 3.88 (t, J = 1 1.8 Hz, 2H), 2.27 (dt, J = 13.3, 3.1 Hz, IH), 2.15 - 2.06 (m, IH). LCMS~ESI+ (ni/z): [M+H calculated for C21H20F2N3O6: 448.40; found: 448.2.
Ste 3b
Magnesium bromide (13 mg, 0.07 mmol ) was added to a solution of intermediate 51-A (13.2 mg, 0.03 mmol, absolute stereochemistry unknown) in 1 mL acetonitrile. The reaction mixture was stirred at 50 °C for 1 hour, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgS04, filtered, concentrated, and purified by HPLC (ACN H20 with 0.1 % TFA modifier) to afford compound 51. 1 H-NMR (400 MHz, CMoroforn ) δ 10.47 (t, J 5.8 Hz, I H), 8.42 (s, IH), 7.35 (q, 8.6, 8.2 Hz, I H), 6.81 (q, J = 8.7, 8.0 Hz, 2H), 6.41 (dd, J = 10.0, 3.6 Hz, IH), 4.79 (s, IH), 4.65 (s, 2H), 4.36 - 4.26 (m, 21 1 ). 4.20 - 4.08 (m, 2H), 3.98 (dd, J = 12.4, 10.2 Hz, I H), 3.88 (t, J = 1 1.8 Hz, 2H), 2.27 (dt, J = 13.3, 3.1 Hz, IH), 2.15 - 2.06 (m, IH). LCMS~ESI+ (ni/z): [M+H calculated for C21H20F2N O6: 448.40; found: 448.2. Example 52
Preparation of Compound 52
(2S,5R,13aS)-N-(2-cyclopropoxy-4-fluorobenzyl)-8-hydroxy-7,9-dioxo- 2,3,4,5,7,9,13 ,13a-octahydro-2,5-methanopyrido[ ,2':4,5]pyrazino[2,l - b] [ 1 ,3]oxazepine- 10-earboxamide
Figure imgf000114_0001
Step 1
A solution eyelopropanoi (1 ,9 g, 29 mmol) in 20 ml. dioxane was added dropwise to a 0 °C solution of Sodium hydride (60% dispersion in mineral oil, 1 .04 g, 26 mmol) in 80 mL dioxane. The reaction mixture was allowed to warm to room temperature, 2,4-difluorobenzonitrile (3.48 g, 25 mmol) was added portionwise, and reaction temperature raised to 95 °C. The reaction solution was cooled to room temperature after stirring for 18 hours, diluted with ethyl acetate, washed twice with water and twice with brine, dried over MgS04, filtered, and concentrated onto silica gel. Purification by silica gel chromatography (0-10% EtO Ac/hex an es) afforded 2- cyclopropoxy-4-fluorobenzonitrile. iH~NMR (400 MHz, Chloroform-* ) 6 7.52 (dd, J :::: 8.6, 6.2 Hz, IH), 7.05 (dd, J = 10.5, 2.3 Hz, ! H), 6.73 (id, J = 8.2, 2.3 Hz, 1 H), 3.87 -
3.76 (m, IH), 0.87 (m, 4H).
Step 2
To a 0 CC suspension of lithium aluminum hydride in THF (1M, 15 mL, 15 mmol) was added 2-cyclopropoxy-4-fluorobenzonitrile in 14 mL diethyl ether dropwise. The reaction solution was stirred for 3 hours, gradually warming to room temperature, at which point it was recooled to 0 °C, an additional 8 ml. lithium aluminum hydride in THF (1M, 8 mmol) added, and stirred for an additional 90 minutes. The reaction was quenched by sequential addition of 0.9 mL water, 0.9 mL 15% aOH aq), and 2,7 mL water. The reaction was filtered through celite with diethyl ether rinses, dried over MgS04, and concentrated to afford 2-cyclopropoxy-4- fiuorobenzyl amine of sufficient purity to carry on as crude. 1H-NMR (400 MHz, Chloroform^/) 8 7.17 - 7.08 (m, IH), 6.96 (dd, J = 10.9, 2.4 Hz, 1H), 6.61 (td, J = 8.3, 2.5 Hz, 1H), 3.78 - 3.66 (m, 3H), 0.89 - 0.72 (m, 4H). Step 3
Compound 15-B (46 mg, 0.14 mmol) was taken up in 1 mL acetonitriie and treated with 2-cyciopropoxy-4-fiuorobenzylamine (32 mg, 0, 18 mmol ), HATU (62 mg, 0.16 mmol), N,N-diisopropylethylamine (DIPEA) (0.04 mL, 0.22 mmol), and stirred at room temperature for 2 hours, after which LCMS analysis revealed complete consumption of compound 15~B and formation of intermediate 52~A. The reaction mixture was carried onto the next step.
Step 4
To the cmde reaction solution of the previous step was added MgBr2 (56 mg, 0.30 mmol). The reaction mixture was stirred at 50 °C for 90 minutes, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgS04, filtered, concentrated, and purified by HPLC (ACN/¾0 with 0.1% TFA modifier) to afford compound 52. ^l-N R (400 MHz, DMSO-</6) δ 12.44 (s, I H), 10.21 is. ./ 5.8 Hz, IH), 8.41 (s, ! ! ! }. 7.22 - 7.15 (m, I FF), 7.12 (dd, J - 1 1.2, 2.5 Hz, IH), 6,72 ( id. ./ 8.5, 2.5 Hz, IH), 5.42 (dd. J 9.6, 4.1 Hz, 1 1 1 ). 5.07 (s, I H), 4.66 (dd, J = 12.8, 4.1 Hz, 1H), 4.58 (s, I H), 4.34 (dd, J ------ 5.6, 2.4 Hz, 2H), 4.04 - 3.91 (m,
2H), 1 .92 (s, 4! I ). 1 .82 id. J ------ 1 1 .9 Hz, 1 H), 1.55 (dx, 12.4, 3.5 Hz, 1 H ), 0.80 (q, J ------
6.3, 5.7 Hz, 2H), 0.72 (q, J = 6.0, 4.9 Hz, 2H). LCMS-ESI+ (m/z) | M i ! j calculated for C24H25FN3O6: 470.17; found: 470. 1.
Example 53
Preparation of Compound 53
(2R,5S,13aR)-N-(2-cyciopropoxy-4-fluorobetizyl)-8-hydroxy-7,9-dioxo- 2,3,4,5,7,9, 13, 1 3a-octahydro-2,5-methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 - b][l ,3]oxazepine-l 0-carboxamide
Figure imgf000116_0001
Compound 42-A (46 mg, 0.14 mmol) was taken up in 1 mL acetonitrile and treated with 2-cyclopropoxy-4-fluorobenzylamine (33 mg, 0.18 mmol), HATU (61 mg, 0.16 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.04 mL, 0.24 mmol), and stirred at room temperature for 2 hours, after which LCMS analysis revealed complete consumption of compound 42-A and formation of intermediate 53~A. The reaction mixture was carried onto the next step. Step 2
To the crude reaction solution of the previous step was added MgBr2 (55 mg, 0.30 mmol). The reaction mixture was stirred at 50 °C for 90 minutes, acidified with 10% aqueous HCl, partitioned between the aqueous and dichioromethane, and the aqueous phase extracted with dichioromethane. The combined organic phases were dried over MgS04, filtered, concentrated, and purified by HPLC (ACN/¾0 with 0.1% TFA modifier) to afford compound 53. 1H~NMR (400 MHz, DMSG-<i6) δ 12.44 (s, 1H), 10.21 (t, J= 5.8 Hz, 1H), 8.41 (s, III .7.22 - 7.15 (m, 1H), 7.12 (dd, J= 11.2, 2.5 Hz, 1H), 6.72 (id,./ 8.5, 2.5 Hz, 1H), 5.42 (dd../ 9.6, 4.1 Hz, III .5.07 (s, 1H), 4.66 (dd, ./ 12.8, 4.1 Hz, 1H), 4.58 (s, 1H), 4.34 (dd. J = 5.6, 2,4 Hz, 2H), 4.04 - 3.91 (m, 2H), 1.92 (s, 4H), 1.82 id. J 11.9 Hz, IH), 1.55 id;../ 12.4, 3.5 Hz, 1H), 0.80 (q../ 6.3, 5.7 Hz, 2H), 0.72 (q, J= 6.0, 4.9 Hz, 2H). LCMS-ESF (jn/z): [M+Hf calculated for C24H25FN3O6: 470.17; found: 470.1.
Example 54
Preparation of Compound 54
(2R,5S)-N~((S)-l~(2,4~difluorophenyl)-2,2,2 xifluoroethyi)-8
2,3,4,5,7,9,13,13a-octahydro-2,5-rnethanopyrido[ ,2':4,5]pyrazino[2, 1- b] [ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000117_0001
Figure imgf000118_0001
54
Step 1
A 50-mL round bottom flask was charged with reactant 54-A (0.02 g, 0.06 mmol), (S)-l-(2,4-difluorophenyl)-2,2,2-trifluoroethanamine (0.019 g, 0.09 mmol), RN-diisopropylethylamine (DIPEA) (0.048 g, 0.38 mmol) and HATU (0.036 g, 0,09 mmol) in DCM (2 ml). The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated down, re-dissolved in EtOAc (50 mL), washed with saturated NaHCOj (2x), saturated NH4CI and dried over Na2S04. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to obtain 54-B. LCMS-ESI+ (m/z): \ VI - H i calculated for i VJ I i J-' s O-: 514; found: 514.
Step 2
A 50-mL round bottom flask was charged with reactant 54~B (0.03 g, 0.058 mmol) and magnesium bromide (0.03 g, O. lSmmol) in acetonitrile (2 mL). The reaction mixture was heated to 50 °C, After 10 minutes, the reaction mixture was cooied to 0 °C and I N hydrochloric acid (0.5 mL) was added in. Then the reaction mixture was diluted with MeO (2 mL). After filtration, the crude was purified by Pre- HPLC purification (30-70% aeetonitrilerwater, 0.1% TFA) afforded compound 54 as TEA salt. 1H-NMR (400 MHz, Chloroformed) δ 11.28 (d, J = 9.4 Hz, 1H), 8.39 (s, 1H), 7.54 (q, J = 7.8 Hz, 1H), 7, 12 - 6.76 (m, 2H), 6.40 - 5.98 (m, 1H), 5.57 - 5.18 (m, 2H), 4.68 (s, 1H), 4.29 (dd, J = 13.1, 4.0 Hz, 1H), 4,05 (dd, J = 12.9, 9.3 Hz, 1H), 2.39 - 1.94 (m, 4H), 1 .86 (t, J = 10.5 Hz, 1 H), 1.60 (dt, J - 12.6, 3.4 Hz, 1 H). 19F-NMR (376 MHz, Chloroform-d) δ -75.30 (t, J = 6.8 Hz, 3 F), -108,33 (dd, J = 8.6, 6.3 Hz, IF), -111.56 - - 13,23 (m, 1 F). LCMS-ESf (m/z): [M i I i] calculated for C21H 0F2N3O5 : 500.; found; 00,
Example 55
Preparation of Compound 55
(lR,4SJ2aS)-7-hydroxy-6 -dioxo-N-(2,4,6-trifluorobenzyl)-i>23,4>6^ 2,12a- octahydro-l,4-metlianodipyrido[L2~a:r,2'-d]pyrazine-9-carboxarnide
Figure imgf000119_0001
Figure imgf000119_0002
55· 55-F
Figure imgf000119_0003
55-G 55-H
Figure imgf000119_0004
Step 1
A mixture of compound 55- A (40.60 g, 150 mmol) and Pd(OH)2/C (12 g) in EtOH (400 mL) under an atmosphere of Ha was stirred at room temperature overnight. The reaction mixture was filtered and treated with HCI/EtOH (400 mi). The mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated to give compound 55-B, which was used in next step without purification. LCMS- ESI+ (m/z): [M+H] ; calculated for C9H16NO: 170.1.; found: 170.2. Step 2
To a solution of compound 55-B (92.25 g, 0.45 mol) and 2CO3 ( 1 86.30 g, 1.35 mol) in ( Ί hC \ (1 L) was added benzyl bromide (76.50 g, 0.45 mol) at 0 °C. The mixture was stirred at room temperature overnight. The reaction mixture was filtered, concentrated and the residue was purified by chromatography on silica gel to give compound 55-C.
Step 3
To a mixture of diisopropylamme (50 g, 0,50 mol) in THF (400 mL) was added n-BuLi (200 mL, 0.50 mol) at -78 °C at N2 atmosphere. After 0.5 h, the reaction mixture was warmed to 20 °C and stirred for 0.5 h. The mixture was cooled to -78 °C and added a solution of compound 55-C (64.75 g, 0.25 mol) in THF (600 mL) under N? atmosphere. The mixture was stirred for 4 h and quenched with saturated NH4Cl solution. The mixture was extracted with EtOAc and the organic layer was washed with brine, dried over Na2S04, filtered and concentrated. The residue was purified by chromatography on silica gel to give compound 55-D.
Step 4
A mixture of compound 55~D (129.50 g 0.50 mol) in 4N HO (1.30 L) was refluxed for 4 h. the mixture was concentrated. The residue was purified by HPLC to gi ve compound 55-E. Step 5
To a mixture of compound 55~E (47 g, 176 mmol) and Pd(OH)2/C (9 g) in EtOH (400 mL) under an atmosphere of H2 was stirred at room temperature overnight. The reaction mixture was concentrated to give compound 55-F, which was used in next step without purification. !H-NMR (400 MHz, CDC13) δ 4.22 (s, I H), 4.06 (s, i l l ). 2.98-2.95 (d. ./ 1 1.2 Hz, 1 H), 1 .96-1 .93 id. 1 1.2 Hz, IH), 1.86-1.82 (m, 2H), 1.76-1.74 (d, J = 9.2 Hz, 2H), 1.49 (s, IH). LCMS~E8I+ (m/z): [ - H j calculated for C7H12NO2: 142.1 .; found: 142.1 .
Step 6
To a mixture of compound 55-F (29.20 g, 165 mmol) and 2 aOH solution (330 mL, 0.66 mol) in dioxane (120 mL) was added Boc20 (39.60 g, 181 mmol) at 0 °C. The reaction mixture was stirred at room temperature overnight. The mixture was adjusted with 3N HCl to pH::::5~6 and extracted with DCM. The organic layer was dried over Na2S04, filtered and concentrated to give 55-G. 1H-NMR (400 MHz, CDCI3) δ 4.40 (s, I H), 4.26 (s, I H), 2.89 (s, 1 H), 1 .76-1 .74 (s, IH), 1.69-1.59 (m, 4H), 1.50 (s, IH), 1.47 (s, 9H). LCMS-ESI* (m/z): [M+Na calculated for Ci 2H19NNa04: 264.1.; found: 264.1.
Step ?
To a mixture of compound 55-G (500 mg, 2.07 mmol) in THF (10 mL) chilled to 0 °C was added BH3-DM8 THF complex (2N in THF, 8.23 mmol, 4.1 ml.) slowly. Gas evolution occured. Internal temperature was monitored to ensure no major exotherm. Reaction was allowed to warm to r.t. overnight. Some starting material remained by LC/MS, additional 2 mL BH3-DMS THF complex was added and the mixture was stirred for additional 3 hr then cooled reaction to 0 °C and slowly quenched with methanol (gas evolution occurs). Internal temperature monitored to ensure exotherm below 25 °C. The mixture was concentrated then purified by silica gel chromotography (20-40% EtOAc/Hexanes) to afford 55-H. Step 8
Compound 55 was prepared as described for Example 41, substituting 55-H for 41-B to afford compound 55. 1H-NMR (400 MHz, DMSO-d6) δ 11.81 (s, IH), 10.40 (t, J = 5.8 Hz, 1H), 8.39 (s, 1H), 7.19 (t, J = 8.6 Hz, 2H), 4.59 - 4.48 (m, 4H), 4.16 ft, J = 12.2 Hz, IH), 4.03 (d, J = 12.2 Hz, IH), 2.69 (s, ill).1.75 (d, J - 10.1 Hz, IH), 1.69 - 1.55 (m, 5H).19F NMR (376 MHz, DMSO-d6) δ -109.3 (m, IF), -112.5 (m, IF). LCMS-ESf (m/z): [M+E]+ calculated for C21H19F3 3O4: 434.13.; found: 434.32.
Example 56
Preparation of Compound 56
(l ,2S,4 ,12aR.)-2-fj.uoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzy[)- l,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a:l 2'-d]pyrazine-9- carboxamide
Figure imgf000122_0001
Figure imgf000123_0001
5S-A 56-B 56-C
Figure imgf000123_0002
Step 1
A solution of 56-A (5 g, 19.43 mmoi) in tetrahydrofuran (65 ml) was cooled in an ice bath as 0.5 M 9-borabicyclo[3.3.1 ]nonane (48.58 ml) was added dropwise. The reaction mixture was warmed up to room temperature. After 18 hours, the reaction wras cooled to 0 °C and a mixture of 2M sodium hydroxide (34 ml) and hydrogen peroxide (9.34 ml, 97.15 mmoi) was added dropwise. After 2 hours at 0 °C, the reaction was warmed up to room temperature and stirred for 1 hour. The mixture was diluted with EtOAc and washed with water. The aqueous fractions were extracted with EtOAc, and the organic fractions combined were dried (Na2S04) and concentrated. The residue was purified by silica column chromatography (50-70% EtOAc/hexanes) to afford 56-B (3.05 g, 57%). LC S-ESI+ (m/z): [M+Hf calculated for C :,.H. - i N .>: 275.34; found: 276.122. Step 2
To a solution of 56-B (1 .45 g, 5.27 mmol) in N,N~dimethylforaiamide (12 ml) was added tert-butyichlorodiphenylsilane (1 .51 ml, 5,79 mmol) and imidazole (1.08 g, 15.8 mmol). After 18 hours, the mixture was diluted with water, extracted into EtOAc (2x), the organic fractions were combined, dried (Na2S04), and concentrated. The residue was purified by silica column chromatography (10-20% EtOAc/'hexanes) to afford 56-C (2.6 g, 96.1%). LCMS-ESI+ (m/z): [M+H] calculated for C :..I \0.<Si : 513.74; found: 514.625.
Step 3
To a solution of 56-C (3.27 g, 6.36 mmol) in EtOH (26 mL) and acetic acid (3 mL) was added 10% PdOH/C (0.52 g, 3.7 mmol) and the suspension was shaken in a Parr apparatus at 50 atm for 20 hours. After filtering through Celite, the cake was washed with EtOH, the filtrate was concentrated under vacuum. The residue was dissolved in ethanol (26 ml) and acetic acid (3 ml, 52.4 mmol), treated with 10% PdOH/C (0.52 g, 3.7 mmol) and shaken in a Parr apparatus at 50 atm for 20 hours. Filtered through Celite, the cake was washed with EtOH, the filtrate was concentrated under vacuum to dryness to afford the crude deprotected product (2.07g, 79.4 %). LOIS- ESI (m z): i . i · H | calculated for ( · Η « , \Ό.^ΐ : 409.59; found: 410.485.
To the crude residue (2 g, 4.88 mmol) and di-tert-butyl dicarbonate 97%> (2.14 g, 9.79 mmol) in THF (20 ml) was added N,N-diisopropylethylamine (DIPEA) (2.14 ml, 12.27 mmol). After 20 h, the reaction mixture was diluted with water, extracted into EtOAC (2x) and the two organic fractions were washed with water, combined, dried (Na2S04), and concentrated. The residue was purified by silica column chromatography (10-20% EtOAc/Hexanes) to afford 56-D (2.1 3 g, 86.14%). LCMS- ESI+ (m/z): [M+H]" calculated for C3oH41N05Si: 523.74; found: 523.922.
Step 4
A solution of 56~D (2.07 g, 4.06 mmol) in THF (20 mi) was stirred in an ice bath as 2.0 M LiBH4 in THF (4.07 ml) was added and the resulting mixture was stirred at room temperature for 18 h. After, the reaction mixture was diluted with ethyl acetate and treated slowly with water. The two phases were separated, and the aqueous fraction was extracted again with ethyl acetate. The two organic fractions were washed with water, combined, dried ( a2S04), and concentrated. The residue was purified by silica column chromatography (20-40% EOAc/hexanes) to afford 56-E (1 .59 g, 81 .3%). LCMS-ES (m/z): j VI · H | calculated for C .J h-.Λ ;Si : 481.7; found: 482.337.
Step 5
A mixture of 56-E (1.58 g, 3.28 mmol), phthaliraide (0.79 g, 5.38 mmol) and triphenylphosphine (1.93 g, 7.37 mmol) in THF (90 ml) was cooled in an ice bath. Diisopropyl azodicarboxylate, 95% (1 .46 ml, 7.42 mmol) was added. The mixture was then warmed up to room temperature and stirred for 20 h. After, the reaction mixture was concentrated and the residue dissolved in ether, cooled in an ice bath and stirred for 1.5 h. The solids were filtered off and the filtrate was concentrated. The residue was purified by silica column chromatography (10-30% EtOAc/hexanes) to afford the protected amino compound (1.86 g, 92.8%).
A solution of the protected amino compound 56~F (1.85 g, 3.03 mmol) and hydrazine hydrate (0.6 ml, 12.39 mmol) in ethanol (19 ml) was stirred at 70 °C for 2 h. The reaction mixture was cooled in an ice bath, ether (10 ml) was added and the mixture was stirred for 30 mm. The solid formed was filtered off and the filtrate was concentrated under vacuum to ryness.
Step 6
A mixture of crude amino compound 56-F (991 mg, 2.06 mmol), compound 38-F (Example 38) (714 mg, 2.06 mmol) and NaHC03 (347 mg, 4.12 mmol) in water (15 niL) and EtOH (15 mL) was stirred for 20 h. The reaction mixture was concentrated under vacuum and the residue was partitioned between water and EtOAc. The aqueous layer was re-extracted with EtOAc and the combined organic layers were dried (Na2S04) and concentrated. The residue (1.5 g) was dissolved in CH2CI2 (5 mL) and 4N HC1 in dioxane (18.6 mL) was added. After 1.5 hours the mixture was concentrated to dryness, co-evaporated with toluene and dried in vacuo.
The crude residue (1.38 g) and DBU (1.4 ml, 9.38 mmol) in toluene (25 ml) was stirred at 1 10 °C. After 35 minutes the mixture was concentrated and the residue was purified by silica column chromatography (5-15% MeOH EtOAc) to afford 56~G (450 mg, 72,3%), LCMS-ES (m/z): [M+HJ+ calculated for (\,l [ |.>VO{1Si : 662.85; found: 663.766. Step 7
The mixture of 56-G (890 mg, 1.34 mmol) in MeOH (14 ml) and THF (14 ml) was stirred at room temperature as 1M OH (7.09 ml) was added. After 30 min the reaction mixture was neutralized with IN HQ, extracted into EtOAc (2x) and the combined organic extracts were dried (Na2S04) and concentrated.
A suspension of the cmde residue (850 mg), 2,4,6-trifluorobenzylamine
(248 mg, 1.54 mmol ) and HATU (662 mg, 1.74 mmol) in dichioromethane (5 ml) was stirred at room temperature as Ν,Ν-diisopropylethylamine (DIPEA) (1 .63 ml, 9.37 mmol) was added. After 1 h, additional 2,4,6-difluorobenzylamine (32 mg, 0.2 mmol), HATU (153 mg, 0.4 mmol) and Ν,Ν-diisopropylethylamine (DIPEA) (0.12 ml, 0.67 mmol) were added. After 30 minutes the mixture was diluted with water, extracted into EtOAc (3x) the combined organic phases were dried (Na2S04), concentrated and the residue was purified by silica column chromatography (50-75% EtOAc/hexanes) to afford 56- I f (919 mg, 88.23%). LCMS-ESI+ (m/z): i VI · 1 1 1 calculated for C - i - i S:: 777.9; found: 778.409.
Step 8
A solution of 56-H (915 mg, 1.18 mmol) in THF (5 ml) was stirred in an ice bath as 1.0 M tetrabutyl ammonium fluoride in THF (1 , 18 ml) was added dropwise. The resulting mixture was stirred at room temperature for 30 min. The reaction mixture was concentrated under vacuum and the residue was diluted with EtOAc, washed with water, dried ( a2S04), concentrated and the residue was purified by si lica column chromatography (50-75% EtOAc hexanes then 5% MeOH/EtOAc). The resulting material (248 mg, 0.46 mmol) was dissolved in dichioromethane (2 ml) cooled to -78°C as diethylaminosulfur trifluoride (0.07 mL, 0.55 mmol) was added dropwise and the reaction was warmed to room temperature and stirred for 1 h. The reaction was cooled in an ice bath and quenched with saturated NaHC03, two phases were separated, and the separated aqueous fraction was extracted with CH2C12. The two organic fractions were combined dried (Na2S04), and concentrated. The residue was purified by silica column chromatography (1% MeOH/EtOAc) to afford 56-J (75 mg) (LCMS-ESF (ni/z): [M+H]+ calculated for C28H23F4N3O4 : 541.49; found: 542.320) and 56-1 (30 mg) (LC S-ESI+ ijn/z): [M+H]+ calculated for ( '..xI -W^O i : 521 .49; found: 522.05).
Compound 56-J (75 mg, 139 mmol) was dissolved in TFA (1 iriL), stirred at room temperature for 10 minutes, and the solution was concentrated. The residue was purified by reverse phase HPLC (Gemini, 15 to 43% ACN/H20 + 0.1 % TFA) to afford compound 56. ^-NMR (400 MHz, DMSO- 6) δ 10.67 (s, I H), 7.80 (s, IH), 7.17 (t, J = 8.6 Hz, 2H), 5.45 - 5.18 (m, IH), 4.70 - 4.39 (m, 3H), 4.23 (d, J i 1 .5 Hz, I H), 4.1 1 - 3.85 (m, 2H), 2.85 (dd, J = 4.2, 2.0 Hz, H), 2.34 - 2.13 (m, I H), 1.81 (s, IH), 1.55 - 1.33 (m, 2H). "F-NMR (376 MHz, DMSO-ifc) δ -74.20 (m), - 106.95 - -1 16.45 (m), -190.65 - -194.54 (m).
Example 57
Preparation of Compound 57
(lR,4R., 12aR)-2,2-difluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifiuorobenzyi)- l ,2,3,4,6,8, 12, 12a-octahydro- 1 ,4-methanodipyrido[ 1 ,2-a: 1 ',2'-d]pyrazme-9- carboxamide
Figure imgf000127_0001
57
Figure imgf000128_0001
57-E 57-F
TF:A
Figure imgf000128_0002
57-G 57-H
Figure imgf000128_0003
57
StepJ.
A solution of 57-A (1.45 g, 5.34 mmol) in dichloromethane (30 ml) was cooled in an ice bath as Dess Martin periodinane (4.53 g, 10.69 mmol) was added in portions and the reaction was stirred at room temperature for 18 h. The reaction was quenched by addition of water, the precipitate was filtered off and a saturated solution of Na2S203 was added. The mixture was stirred until the triphasic solution turned then saturated NaHC03 was added and the aqueous layer extracted with CH2CI2. The combined organic fractions were dried (Na2S04) and concentrated. The residue was purified by silica column chromatography (30-50% EtOAc/Hexanes) to afford 57-B (1.13 g, 78.2 %). LCMS-ESr (ni/z) [M+Hf calculated for C0H19NO5: 269.29; found: 269.722.
Step 2
A solution of 57~B (0.5 g, 1.86 mmol) in dichloromethane (10 ml) was cooled to -78°C as dietbyiammosulfur trifhioride (0.52 mL, 3.91 mmol) was added dropwise and the reaction was warmed to room temperature and stirred for 18 h. The reaction was cooled in an ice bath and quenched with saturated NaHCQ3, two phases were separated, and the separated aqueous fraction was extracted with (Ί h('k The two organic fractions were combined, dried (Na2S04) and concentrated. The residue was purified by silica column chromatography (20-50% EtOAc/hexanes) to afford 57-C (518 mg, 95.39%). !H-NM (400 MHz, Chloroform-d) δ 4.43 (s, 1H), 4.36 - 4.27 (m, 1H), 4,22 (s, 1H), 3.75 (s, 3H), 2.95 (t, J = 8.1 Hz, 1H), 2.30 - 1.98 (m, 2H), 1.85 - 1.71 (m, 1H), 1.44 (m, 9H).
Step 3
A solution of 57-C (935 mg, 3.21 mmol) in THF (10 ml) was stirred in an ice bath as 2.0 M LiBH4 in THF (3.22 ml) was added and the resulting mixture was stirred at room temperature for 18 h. After, the reaction mixture was diluted with ethyl acetate and water was added slowly. The two phases were separated, and the separated aqueous fraction was extracted with ethyl acetate. The two organic fractions were washed with water, combined, dried (Na2S04), and concentrated. The residue was purified by silica column chromatography (20-40% EtOAc/hexanes) to afford 57-D (724 mg, 85.67%). ^l-NMR (400 MHz, Chloroform-*/) δ 4.30 - 3.48 (m, 5H), 2.75 - 2,56 (m, 1 H), 2.24 - 1.90 (m, 3H), 1 .86 - 1 .65 (m, 1 H), 1.47 (s, 9H). Step 4
A mixture of 57-D (720 mg, 2.74 mmol), phthalimide (402 mg, 2.73 mmol) and triplieiiylpliosphine (1.61 g, 6.15 mmol) in THF (45 ml) was cooled in an ice bath. Diisopropyi azodicarboxylate, 95% (1.22 ml, 6.19 mmol), was added. The mixture was then warmed up to room temperature and stirred for 20 h. After, the reaction mixture was concentrated and the residue dissolved in ether, cooled in an ice bath and stirred for 1.5 h. After the solids were filtered off, the filtrate was concentrated. The residue was purified by silica column chromatography (40-60% EtOAc/hexanes) to afford the phthalimide adduct (1.07 g, 99.7%). LCMS-ESI+ (m/z): [M+Hf calculated for C2oH22F2N204: 392.4; found: 393.204
A solution of the phthalimide adduct (1.07 g, 2.73 mmol) and hydrazine hydrate (0.54 mL, 11.15 mmol) in etbanol (10 ml) was stirred at 70 °C for 2 hours. The reaction mixture was cooled in an ice bath and ether (10 ml) was added. The mixture was stirred for 30 min. The solid formed was filtered off and the filtrate was concentrated under vacuum to dryness to afford crude 57- E.
Step 5
A mixture of crude 57-E (709 mg, 2.7 mmol) compound 38-F (Example
38) (936 mg, 2.7 mmol) and NaHC03 (454 mg, 5.41 mmol) in water (15 mL) and EtOH (15 mL) was stirred for 20 h. The reaction mixture was concentrated under vacuum and the residue was partitioned between water and EtOAc. The aqueous layer was re- extracted with EtOAc and the combined organic layers were dried (Na2S04) and concentrated. The residue (1.5 g) was dissolved in CH2CI2 (7 mL) and 4N HC1 in dioxane (26.9 mL) was added. After 1.5 hours the mixture was concentrated to dryness, co-evaporated with toluene and dried in vacuum. The crude residue (1.3 g) and DBU (2 ml, 13.4 mmol) in toluene (25 ml) was stirred at 1 10 °C. After 35 minutes the mixture was concentrated and the residue was purified by silica column chromatography (5- 15% MeOH/EtOAc) to afford 57~F (426 mg, 36.17%). LCMS-ESI+ (m/z): | M H i calculated for C nH -F..\ ><).:: 444.43; found: 445 ,280,
Step 6
The mixture of compound 57-F (426 mg, 0.96 mmol) in MeOH (7 ml ) and THE (7 ml) was stirred at room temperature as 1M KOH (5.06 ml) was added. After 30 minutes the reaction mixture was neutralized with IN HQ, extracted into EtOAc (2x) and the combined organic extracts were dried (Na2S(>4) and concentrated to crude 57-G. Step 7
A suspension of the crude residue 57-G (189 mg), 2,4,6- trifluorobenzylamine (95 mg, 0.59 mmol) and HATU (276 mg, 0.73 mmol) in dichloromethane (3 ml) was stirred at room temperature as N,N-diisopropylethylamine (DIPEA) (0.59 ml, 3,4 mmol) was added. After 1 h he mixture was diluted with water, extracted into EtOAc (3x). The combined organic phases were dried (Na2S04) and concentrated to 57-H. LCMS-ESI* (m/z): [M+H]+ calculated for C28H22F5N3O4: 559,48; found: 560.24, Step 8
Compound 57-H (150 mg, 0.27 mmol) was dissolved in TFA (2 mL), stirred at room temperature for 10 min, and the solution was concentrated. The residue was purified by reverse phase HPLC (Gemini, 15 to 60% ACN/H20 + 0.1% TFA), to afford compound 57 (85 mg, 67.5%). LCMS~ES (m/z): [M+H]+ calculated for C2JH16F5N3O4: 469.36; found: 470.229. iH-NMR (400 MHz, OMSO-d6) δ 10.41 (t, J = 5.6 Hz, I H), 8.20 (s, I I I ). 7.12 (t, J -- 8.7 Hz, 2H), 4.79 (s, I H), 4.48 (m, 3H), 4.10 (m, 2H), 3.02 (d, J ------ 5.7 Hz, IH), 2.33 (m, IH), 2.22 - 1.97 (m, 2H), 1.85 (d, J ------ 11.0 Hz, I H), 1 .21 (s, IH). ; '·'" NMR. (376 MHz, DMSO-</6) δ -69.88 , -71.77 , -74.09 , -88.33 ( ld, ./ 222.6, 23.8 Hz), -109.15 - -109.60 (m), -110.04 , -1 12.44 (K ./ 7.6 Hz).
Example 58
Preparation of Compound 58
(lR,4RJ2aR)-N-(3-cWoro-2,4-difluoroberiz d)-2,2-difiuoro-7-hydroxy-6,8-dioxo- 1 ,2,3,4,6,8, 12,12a-octahyd.ro- 1 ,4-methanodipyrido[ 1 ,2~a: .1 ',2'-d]pyrazine-9- carboxami.de
Figure imgf000131_0001
58 Step 1
A suspension of the crude residue 57-G (120 mg), 3~chloro,2,4- difluorobenzylamine (67 mg, 0.38 mmol) and HATU ( 175 mg, 0.46 mmol) in dichloromethane (3 ml) was stirred at room temperature as N,N-diisopropylethylamine (DIPEA) (0.38 ml, 0.28 mmol) was added. After 1 h the mixture was diluted with water, extracted into EtOAc (3x) the combined organic phases were dried (Na2S04) and concentrated to yield 58-A. LCMS-ESI+ (m/z) [M+H]+ calculated for C28H22QF4 3O4: 575.94; found: 576.394.
Step 2
Compound 58-A (166 mg) was dissolved in TFA (2 mL), stirred at room temperature for 10 min, and the solution was concentrated. The residue was purified by reverse phase HPLC (Gemini, 15 to 70% ACN/H20 + 0.1% TFA), to afford compound 57 (60 mg, 42.8%). LCMS-ESI+ (m/z): [M+H] calculated for C21H16CIF4N3G4: 485.82; found: 486.135. ¾- MR (400 MHz, DMSO-d6) δ 10.77 (t, J = 6.0 Hz, 1H), 7.77 (s, 1 1 1 ), 7.28 (m, 2H), 4.77 (s, HI), 4.64 - 4.40 (m, 2H), 4.27 (d, J = 9.1 Hz, III), 3.93 (m, 2H), 2.95 i d. J = 5.8 Hz, I H), 2.51 (s, 1 H), 2.42 - 2.17 (m, IH), 2.14 - 1 .89 (m, 2H), 1.77 (m, I H ). 19F-NMR (376 MHz, DMSO-< 6) δ -87.63 , -88.23 , -108.67 , - 109.27 , -1 16.42 (t, ,/ 7.0 Hz), -1 18.48 (d, 7.8 Hz).
Figure imgf000132_0001
Preparation of Compound 59
(] R,2R,4R, 12aR)-2-fluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)- 1 , 23,4,6,8, 12 J.2a-octahydro- 1 , 4-memanodipyri.do[ 1 ,2-a: ,2'-d]pyrazine-9- carboxaniide
Figure imgf000132_0002
59
Figure imgf000133_0001
Figure imgf000133_0002
Step 1
A solution of 57~B (1.9 g, 7.06 mmol) in methanol (35 mL) was stirred at 0 °C as sodium borohydride (667 mg, 17.64 mmol) was added portionwise and the resulting mixture was stirred at room temperature for 30 min. The reaction mixture was cooled in an ice bath, quenched by addition of water and concentrated. The residue was partitioned between water and EtOAc. The aqueous layer was re-extracted with EtOAc and the combined organic layers were dried (Na?SO,¾) and concentrated The residue was purified by silica column chromatography (30-60% EtOAc/hexanes) to afford 59-A (1.49 g). 1H~NMR (400 MHz, chloroform-a') 6 4.57 (s, IH), 4.52 - 4.42 (m, 2H), 4.28 (s, IH), 4.14 (s, IH), 3.72 (d, J = 2.1 Hz, 3H), 2.74 (s, IH), 2.08 - 1.87 (m, 2H), 1.43 (d, J = 23.1 Hz, 10H) and 57-A (96 mg): 1H~ MR (400 MHz, chloroform- /) δ 4.65 - 4.40 (m, 2H), 4.34 - 4.02 (m, IH), 3.73 id. J = 2.3 Hz, M Y). 2.1 A (t, J - 5.3 Hz, I H), 2.12 - 1.55 (m, 3H), 1 .52 - 1.1 8 (m, 1 1 H).
Step 2
To a solution of 59-A (686 mg, 2.53 mmol) in N,N-dimethyiformamide (5 ml) was added tert-butylchlorodiphenylsilane (0.723 mL, 2.78 rrnno!) and imidazole (516 rag, 7.56 rnrnoi). After 18 h, the mixture was diluted with water, extracted into EtOAc (2x), and the organic fractions were combined, dried (Na2S04), and concentrated. The residue was purified by si lica column chromatography (10-20% EtOAc/hexanes) to afford 59-C. LCMS-ESI+ (m/z): | Vi Π ] calculated for C29H39N05Si: 509.71 ; found: 510.793.
Step 3
A solution of 59-C (1.23 g, 2.41 mmol) in THF (13 ml) was stirred in an ice bath as 2.0 LiBH4 in THF (2.42mL, 4.84 mmol)) was added and the resulting mixture was stirred at room temperature for 18 h. After the reaction mixture was diluted with ethyl acetate water was added slowly, two phases were separated, and the separated aqueous fraction was extracted with ethyl acetate. The two organic fractions were washed with water, combined, dried (Na2S04), and concentrated. The residue was purified by silica column chromatography (20-40% EtOAc hexanes) to afford 59-D. LC S~ES (m/z): | VI · ! ! | calculated for (\,J NO iSi : 481.7; found: 482.741.
Sterj_4
A mixture of 59-D (963 mg, 2.0 mmol ), phthalimide (482 mg, 3.28 mmol) and triphenylphosphme (1.18 g, 4.49 mmol ) in THF (50 ml) wras cooled in an ice bath. Diisopropyl azodicarboxylate, 95% (0.89 mL, 4.52 mmol) was added. The mixture was then warmed up to room temperature and stirred for 20 h. After, the reaction mixture was concentrated and the residue dissolved in ether, cooled in an ice bath and stirred for 1.5 h. After, the solids were filtered off and the filtrate was concentrated. The residue was purified by silica column chromatography (10-30% EtOAc/hexanes) to afford the phthalimide adduct. LCM8-ESi+ (m/z): [M+H]+ calculated for P VO--Si : 610.81 ; found: 61 1.935.
A solution of the phthalimide adduct (1.2 g, 1.97 mmol) and hydrazine hydrate (0.4 ml, 8.03 mmol) in ethanol (12 ml) was stirred at 70 °C for 2h. The reaction mixture was cooled in an ice bath and ether (10 ml) was added, the mixture was stirred for 30 min. The solid formed was filtered off and the filtrate was concentrated under vacuum to dryness to afford 59-E. LCMS-ESI+ (m/z) [M+H]4 calculated for C28H4o 203Si: 480.71 ; found: 481.356.
Sterj_5
A mixture of crude 59- E (770 mg, 1.60 mmol), compound 38-F (Example 38) (555 mg, 1.60 mmol) and NaHC03 (269 mg, 3.20 mmol) in water (12 mL) and EtOH (12 mL) was stirred for 20 h. The reaction mixture was concentrated under vacuum and the residue was partitioned between water and EtOAc. The aqueous layer was re-extracted with EtOAc and the combined organic layers were dried (Na2S04) and concentrated.
The residue (1 .29 g) was dissolved in CH2Cl2 (4 mL) and 4N HC1 in dioxane (15.6 mL) was added. After 1.5 hours the mixture was concentrated to dryness, co-evaporated with toluene and dried in vacuum. LCMS-ESI* (m/z): [M+Hf calculated for C n l VO-Si : 708.91 ; found: 709.782.
The crude residue (1.09 mg) and DBU (1.17 ml, 7.8 mmol) in toluene (20 ml) was stirred at 1 10 °C. After 35 min the mixture was concentrated and the residue was purified by silica column chromatography (5-15% MeOH EtOAc) to afford 59- F. LCMS-ESI+ (m/z): ! Vi H j calculated for <\ ί i -X · ·^' " 662.85; found: 663.677. Step 6
A mixture of 59-F (680 mg, 1.03 mmol) in MeOH (10 ml) and THE (10 ml) was stirred at room temperature as IM KOH (5.42 ml) was added. After 30 min the reaction mixture was neutralized with IN HC1, extracted into EtOAc (2x) and the combined organic extracts were dried (Na2SQ4) and concentrated. LC S-ESI4 (m/z):
[M+H]+ calculated for ( I N .O,.Si : 634.79; found: 635.466.
A suspension of the cmde residue (650 mg), 2,4,6-trifluorobenzylamine (214 mg, 1.33 mmol) and HATU (623 mg, 1.64 mmol) in dichloromethane (6 mi) was stirred at room temperature as Ν,Ν-diisopropylethylamine (DIPEA) (1 .34 ml, 7.68 mmol) was added. After 2 h, the mixture was diluted with water, extracted into EtOAc (3x) nad the combined organic phases were dried (Na?S04), concentrated and the residue was purified by silica column chromatography (50-75% EtOAc/hexanes) to afford 59~G. LCMS-ESI+ (m/z): j · H | calculated for ί ' n! ! :..!; .>X ^Si : 777.9; found: 778,566.
Step 7
A solution of 59-G (648 mg, 0.83 mmol ) in THF (10 ml) was stirred in an ice bath as 1.0 M tetrahutylammomum fluoride in THF (0.83 ml) was added dropwise and the resulting mixture was stirred at room temperature for 30 min. Additional 1.0 M tetrabutyiammonium fluoride in THF (0.1 ml) was added dropwise. After 30 minutes, the reaction mixture was concentrated under vacuum and the residue was diluted with EtOAc, washed with water, dried (Na2S04), concentrated and the residue was purified by silica column chromatography (5% MeOH/EtOAc). A solution of the residue (290 mg, 0.54 mmol) in dichloromethane (3 ml)was cooled to -78°C as diethyiaminosulfur trifluoride (0.09 mL, 0.65 mmol ) was added dropwise and the reaction was warmed to room temperature and stirred for 2.5 h. The reaction was cooled in an ice bath, quenched with saturated NaHC0 , two phases were separated, and the separated aqueous fraction was extracted with CH2CI2. The two organic fractions were combined, dried (Na2S04), and concentrated. The residue was purified by silica column chromatography (1% MeOH/EtOAc) to afford 59-H. LCMS-ESI4" (m/z): [ \1 I I I calculated for C >J ί••-1 :4\ !() 1 : 541 .49; found: 542.320. Step 8
Compound 59-H (103 mg, 0.19 mmol) was dissolved in TFA (1.4 mL) at room temperature for 15 min, and the solution was concentrated. The residue was suspended in DMF, filtered off, and the precipitated product was washed with water, dried under vacuum to afford compound 59. LCMS-ESI"' (m/z) [M+H] calculated for <\ | ! ί Γί \\ :<} :: 451 .37, found: 452.226. 'll-NMR (400 MHz, DMSO-d6) δ 1 1 .53 (s, 1 H), 10.35 (t, J = 5.8 Hz, 1 H), 8.34 (s, 1 1 1 ). 7.1 8 (t, J = 8.6 Hz, 2H), 5.15 - 4.88 (m, IH), 4.73 (d, J = 3.3 Hz, 1H), 4.49 (m, 3H), 4.04 (t, J = 12.4 Hz, 1 1 1 ;·. 3.65 (dd, J = 12.4, 3.7 Hz, I H), 2.95 - 2.76 (m, I H), 2.26 - 2.03 (m, lH), 1.96 - 1.64 (m, 3H). 19F-NMR (376 MHz, DMSO- ) 8 -73.93 , -74.74 (d, J = 28.8 Hz), -109.31 (m), -1 12.51 (m), - 165.65 (m). Example 60
Preparation of Compound 60
(l I ,4S,12aR)- ~(23~¾
octahydro-1.,4-methanodipyrido[l ,2-a: ,2'-d]pyrazine-9-carboxamide
Figure imgf000137_0001
Figure imgf000137_0002
60-C
Step 1
To a solution of dimethyl 3-methoxy-4-oxo-4H-pyran-2,5-dicarboxylate (5.5 g, 23 mmol) in MeOH (100 mL) was added 41-E (Example 41) (5 g, 22 mmol) and sodium bicarbonate (3.6 g, 43 mmol). The solution was stirred at room temperature for 1 .5 h. 4M HCl fin dioxane, 55 mL, 221 mmol) was added and the solution was heated to 50 °C for 2h, The reaction was cooled to room temperature and concentrated in vacuo. The resulting oil was dissolved in sodium bicarbonate and washed with EtOAc. The aqueous layers were then extracted with CH2C12 (4x). The combined CH2C12 extractions were dried over Na?S() and concentrated to provide 60-A. LCMS-ESF m/z): | VI - H i calculated for C,,.l l \ >0?: 319.13; found: 319.20.
Step 2
To a suspension of 60-A (3.7 g, 11.6 mmol) in MeOH (12 ml.) and THF (23 mL) was added aqueous K.OH (2M, 15.7 mL, 31.4 mmol). The resulting solution was stirred at room temperature for 10 min. Volatiles were removed in vacuo, and the resulting aqueous layer was acidified with I HCl. The resulting white solid was filtered, washed with water, and dried in vacuo to provide 6Θ-Β. !H-NMR (400 MHz, Chloroformed) δ 8.36 (s, 1H), 5.01 (d, J = 2.7 Hz, 1H), 4.12 (s, 4H), 3.90 (t, J = 12.2 Hz, 1H), 3.78 (dd, J = 12.1, 3.1 Hz, 1H), 2.69 (s, 1H), 1.95 - 1.71 (m, 4H), 1.70 - 1.54 (m, 2H). LCMS-ESI4 (m/z): [M+H]+ calculated for €ί5Η17Ν2θ5: 305.11; found: 305.15.
8tep_J>
To a solution of 60-B (0.10 g, 0.33 mmol ) in CH2C12 (3.5 mL) was added (2,3-diehlorophenyl)methanamine (0.12 g, 0.70 mmol), HATU (0.25 g, 0.66 mmol), and N,N-diisopropylethylamine (DIPEA) (0.29 mL, 1.64 mmol). The resulting solution was stirred at room temperature until judged complete by LC/MS. The reaction mixture was diluted with CH2CI2 and washed with I N HCL The aqueous layer was back-extracted with CH2Ci2, and the combined organic layers were dried over Na2S04 and concentrated in vacuo. The crude material was dissolved in hot DMF and allowed to precipitate upon cooling. Filtration provided 60-C. LCMS-ESI+ (m/z): [M+H]+ calculated for C . -I L -CL O i: 462.10; found: 462.14.
Step 4
To a slurry of 60~C (0.11 g, 0.24 mmol), in acetonitrile (4.5 mL), was added magnesium bromide (0.089 g, 0.48 mmol). The reaction mixture was heated to 45 °C for 2.5 h and then cooled to room temperature. The slurry was diluted with CH2CI2 and washed with IN HCI and brine. The aqueous layers were back-extracted with CH2CI2 (2x) and the combined organic layers were dried over Na2S04 and concentrated in vacuo. The crude solid was triturated with methanoi and filtered to provide 60. Ή-NMR (400 MHz, DMSO- 6) δ 11.72 (s, 1H), 10.50 (t, 1H), 8.34 (s, 1H), 7.55 (dd, 1H), 7.40 - 7.24 (m, 211).4.67 (s, 111}.4.61 (d, 2H), 4.45 (dd, 1H), 3.95 (t, 1H), 3.84 - 3.73 (m, 1H), 1.86 - 1.67 (m, 3H), 1.66 - 1.40 (m, 4H). LCMS-ESI+ (m/z): |\1 111 calculated for ( ..,! !..,( :();: 448.08; found: 448.18.
Example 1
Preparation of Compound 61
(1R,4SJ2aS)-N-(3-ch!oro-2,4-difl^
octahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9-carboxamide
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000140_0002
61 was prepared analogously to Example 60, substituting (1 S,3S,4R)~ tert-b tyl 3-(aminomethy].)-2-azabicyclo[2.2. l]heptane-2-carboxylate (prepared in Example 55) for 41-E, and (3-ch].oro-2,4-dif3.uorophenyl)methanamine for (2,3- dichlorophenyl)methanamine. 'H-NMR (400 MHz, DMSO-d6) δ 1 1.85 (s, 1 H), 10.45 (t, IH), 8.40 (s, I H), 7.37 (td, I I I ). 7.27 (td, IH), 4.63 - 4.46 (m, 4H), 4.17 (t, 1H), 4.04 (dt, 1H), 1.76 (d, IH), 1.73 - 1.54 (m, 5H). LCMS~E8I+ (m/z): ! Vf i { | calculated for C21H19CIF2N3O4: 450.10; found: 450.15.
Preparation of Compound 62
2R,5S, 13aR)-N-(4-fluoro-2-(trifluoromethyl)benzy].)-8-hydroxy-7,9-dioxo- 2 , ,5 ,9J3 J3a-octahydro-2,5-methanopyrido[ ,2':4,5]pyrazino[2,l - b] [ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000141_0001
Compound 62 was prepared in a similar manner to compound 42 using (4-fliioro-2-(tritluorometliyl)p enyl)methanamine in place of (2,4,6- trifulorophenylphenyljmethanamme, iH- M (400 MHz, Chloroform~d) δ 10,50 (s, IH), 8.38 (s, IH), 7.57 (dd, IH), 7.36 (dd, III .7.19 (td, III .5.40 - 5.28 (rn, 2H), 4.79 (t, 2H), 4.69 (s, IH), 4.25 (dd, IH), 4.03 (dd, 1H), 2.17 - 1.98 (m, 4H), 1.96 - 1.84 (rn, 1H), 1.61 (dt, IH). LCMS-ESr in/z): [M+H]+ calculated for Γ.νΙΚ,Γ,Ν,Ο*: 482.13; found: 482.145.
Example 63
Preparation of Compound 63
(2R,5S43aR)-N-(2-chioro-4-fl oroben2yl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13a- octahydro-2,5-metbanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b][ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000141_0002
63
Compound 63 was prepared in a similar manner to compound 42 using (2~chloro~4-fluorophenyl)methanamine in place of (2,4,6- trifulorophenylphenyljmethanamme. iH- MR (400 MHz, Chloroform-d.) δ 10.48 (s, 1H), 8.45 is, IH), 7.39 (dd, IH), 7.12 (dd, IH), 6.93 (td, l!l 5.37 (cL 1 H), 5.31 (t, IH), 4.68 (s, 3H), 4.29 (d, IH), 4.04 (t, IH), 2.21 - 2.01 (m, 4H), 1.97 - 1.82 (m, IH), 1.67 - 1.56 (m, IH). LCMS-ESr (m/z): [M+H]+ calculated for C21H20CIFN3O5: 448.10; found: 448.143. le ί>4
Preparation of Compound 64
(2R,5S J 3aR)~8-hydroxy-7,9~*^
octahydro-2,5-methanopyrido[l\2':4,5]pyrazmo[2,l-b][l,3]oxazepme-10-carboxamide
Figure imgf000142_0001
Compound 64 was prepared in a similar marmer to compound 42 using (2,4,5-trifJuorophenyl)methaiiamine in place of (2,4,6- trifulorophenylphenyl)methananiine. 1H-NMR (400 MHz, Cbloroform-d) 5 10.42 (s, 1H), 8.42 (s, III).7.19 (ddd, III).6.91 (td, III).5.38 (dd, 1H), 5.31 (i 1H), 4.69 (s, 1H), 4.61 (d, 211).4.29 (dd, IH), 4.05 (dd, III).2.18 - 2.02 (m, 41!}. 1.96 - 1.84 (m. IH), 1.66 - 1.56 (m, IH). LCMS~E8F (m/z): [M+H] calculated for ( ,;Π,„1: :() 450.12; found: 450.119.
Example 65
Preparation of Compound 65
(2R,5S,13aR)-N-(5-chloro-2,4-difluorobenzyl)-8-hydroxy
octahydro-2,5-methanopyrido[ 1 ',2':4,5]pyrazmo[2, 1 -b][ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000142_0002
65
Compound 65 was prepared in a similar manner to compound 42 using (5-chloro-2,4-difluorophenyl)methanamine in place of (2,4,6- trifulorophenylphenyljmethanamme. 'H- MR (400 MHz, Chloroform-d) δ 10.47 (t. I H), 8.4.1 (s, I H), 7.40 (dd, IH), 6.90 (t, 1 H), 5.37 (dd, 1H), 5.31 (t, IH), 4.69 (s, IH), 4.62 (d, 2H), 4.28 (d, IH), 4.04 (dd, IH), 2.17 - 2.02 (m, 4H), 1.94 - 1 .86 (m, I H), 1.61 (dt, I H). LCMS-ESr (m/z) | VI · 1 1 1 calculated for C i i ! :>;(Ί Γ .V.O 466.09; found: 466.107. Example 66
Preparation of Compound 66
(lR,4S 2aR)-N-(3^difluorobenzyl)-7½dTOxy-6,8-dioxo ,2,3,4,6,8^2 2a- octahydro-1 ,4-methanodipyrido[ 1 ,2-a: 1 ',2'-d]pyrazine-9-carboxamide
Figure imgf000143_0001
Compound 66 was prepared in a similar manner to compound 60 using (3,4-difluorophenyl)methanamine in place of (2,3-dichlorophenyl)methanamme. XH- NMR (400 MHz, Chloroform^/) δ 10.59 (s, IH), 7.24 - 7.16 (m, 2H), 7.14 - 7.04 (m, 2H), 4.91 (s, IH), 4.58 (d, 3H), 3.94 - 3.82 (m, IH), 3.79 (d, IH), 1.99 - 1.81 (m, 4H), 1.76 (d, IH), 1.70 - 1.60 (m, 3H). LCMS-ESI (m/z): j VI · H | calculated for C2iH2C.F2N304: 416.13; found: 416.415.
Preparation of Compound 67
(lR,4S,12aR)-N-(4-fluoro-2-(trifluoromethyl)benzyl)-7- ydroxy-6,8-dioxo- 1 ,2,3 ,4,6,8, 12, 12a-octahydro- 1 ,4-methanodipyrido[ 1 ,2~a: 1 ',2'-d]pyrazme-9- carboxami.de
Figure imgf000143_0002
67
Compound 67 was prepared in a similar manner to compound 60 using (4-fluoro-2-(trifiuoromethyl)phenyl)methanamme in place of (2,3- dichlorophenyl)methanamine. 1H-NMR (400 MHz, Chloroform-i) δ 11.72 (s, IH), 10.55 (s, IH), 8.29 (s, 1H), 7.61 (s, IH), 7.36 (dd, 1H), 7.18 (id, 1H), 4.91 (s, 1H), 4.80 (d, 3H), 4.11 (s, IH), 1.99 - 1.80 (m, 4H), 1.76 (d, 1H), 1.71 - 1.47 (m, 3H). LCMS- ESI + (m/z): \\\ ill calculated for ( ,·.Ί Ki-V\>0:: 466.13; found: 466.297.
Example 68
Preparation of Compound 68
(lR,4S,12aR)-N-(2-chloro-4-fluorobenzyl)-7-hydroxy-6,8-dioxo- 1,2,3 ,4,6,8, 12, 12a- octahydro-l,4-methanodipyrido[l,2-a: ,2'-d]pyrazine-9-carboxamide
Figure imgf000144_0001
Compound 68 was prepared in a similar manner to compound 60 using (2-chioro-4-fluoropbenyi)methanamine in place of (2,3-dichlorophenyl)methanamine. Ή-NMR (400 MHz, Chloroform-./) δ 11.68 (s, 1H), 10.52 (s, IH), 8.27 (s, 1H), 7.44 - 7.37 (m, IH), 7.11 (dd, III).6.93 (td, IH), 4.90 (s, ill).4.68 (d, 2H), 4.16 - 4.01 (m, IH), 3.88 - 3.70 (m. 211).2.00 - 1.79 (m, 4H), 1.75 (d, IH), 1.70 - 1.57 (m, 2H). LCMS-ESI1 (m/z): [M+H]+ calculated for ( ;Η ,.ίΊΙ- :<);: 432.10; found: 432.214.
Example 69
Preparation of Compound 69
(lR,4S,12aR)-N-(3-chloro-2,4-difluorobenzyl)-7-hydroxy-6,8-d^
octahydro-l,4-metbanodipyrido[l ,2-a: ,2'-d]pyrazine-9-carboxamide
Figure imgf000145_0001
Compound 69 was prepared in a similar manner to compound 60 using (3-chloro-2,4-difluorophenyl)methanamine in place of (2,3- dichlorophenyljmethanamme. 1H-NMR (400 MHz, Chloroform-^/) δ 11.71 (s, 1H), 10.48 (s, 1H), 8.26 (s, ill).7.27 (s, ill).6.92 (td, 1H), 4.90 (s, 1H), 4.66 id.2H), 4.08 (s, 111).3.91 - 3.69 (m, 2H), 2.01 - 1.79 (m, 311). 1.75 (d. 111).1.71 - 1.44 (m, 2H). LCMS-ESI1 (rn/z): [M+H calculated for ,Π,.,ΓΜν θ,: 450.10; found: 450.27. Example 7Θ
Preparation of Compound 70
(lR,4S,12aR)-^
octahydro- 1 ,4-methanodipyrido[ 1 ,2-a: 1 ',2'-d]pyrazine-9-carboxamide
Figure imgf000145_0002
Compound 70 was prepared in a similar manner to compound 60 using (2~i"luoro-3-methylphenyl)methanamine in place of (2,3-dichlorophenyl)methanarmne. 'H- MM (400 MHz, Chloroform-d) δ 11.62 (s, 1H), 10.39 (s, 1H), 8.30 (s, 1H), 7.19 (t, 1H), 7.07 (t, 1H), 6.96 (t, III), 4.89 (d, 1H), 4.67 (d, 2H), 4.08 (s, 1H), 3.88 - 3.67 (m, 2H), 2.26 (cL 3! IK 1.97 - 1.79 (m, 3H), 1.78 - 1.39 (m, 3H). LCMS-ESF (m/z): [VI Hi calculated for C22H23FN3O4: 412.16; found: 412.26. Example 71
Preparation of Compound 71
(lR,4S,12aR)-N-(3,6-dichloro-2-fluoro^
octah dro-1.,4-metbanodipyrido[l ,2-a: ,2'-d]pyrazine-9-carboxamide
Figure imgf000146_0001
Compound 71 was prepared in a similar manner to compound 60 using (3,6-dichloro-2-fluorophenyl)methanamme in place of (2,3- dichlorophenyl)methanamine. 1H- R (400 MHz, Chloroform-d) δ 11.62 (s, 1H), 10.47 (t, 1H), 8.29 (s, ill).7.13 (dd, III).4.88 (s, 1H), 4.85 - 4.73 (m, 2H), 4.09 (d. IH), 3.88 - 3.68 (m, 2H), 1.99 - 1.53 (m, 8H). IX MS- E Si (ni/z): \ VI II] calculated for ,(),: 466.07; found: 466.257. Example 72
Preparation of Compound 72
(l ,4S,12a )-N-(3-chlorobenzyl)-7-hydroxy-6,8-dioxo-l, 2,3,4,6,8,12,12a-octa ydro- 1 ,4-methanodipyrido[ 1 ,2-a: 1 ',2'-d]pyrazine-9-carboxamide
Figure imgf000146_0002
Compound 72 was prepared in a similar manner to compound 6Θ using (3~chiorophenyl)methanamine in place of (2,3-dichlorophenyl)methanarmne. iH- MR (400 MHz, DMSO-d6) δ 11.75 (s, 1H), 10.44 (t, 1H), 8.38 (s, IH), 7.42 - 7.22 (m, 4H), 4.68 (s, ill).4.54 id.2H), 4.48 (dd, IH), 3.97 (t, ill).3.81 (dd, ill).2.58 (s, ill).1.87 - 1.69 (m, 3H), 1.68 - 1.51 (m, 211), 1.46 id, IH). LCMS-ESI''' (m/z): [M+H]'r calculated for C21H21CIN3O4: 414.11; found: 414.21.
Example 73
Preparation of Compound 73
(lR,4S,12aR)-N-i3-chloro-2,6-difluo
octahydro-l,4~methanodipyrido[l,2~a:r,2'-d]pyrazine-9-carboxamide
Figure imgf000147_0001
73
Compound 73 was prepared in a similar manner to compound 60 using (3-chioro-2,6-difluorophenyi)methanamine in place of (2,3- dichioropheiiyl)methanamine. 1 H-NMR (400 MHz, DMSO-d6) δ 11.71 (s, IH), 10.46 (t, IH), 8.34 (s, IH), 7.60 (td, IH), 7.19 (td, IH), 4.67 (s, IH), 4.62 (d, 2H), 4.44 (dd, IH), 3.95 (t, IH), 3.78 (dd, IH), 2.57 (s, IH), 1.86 - 1.68 (m, 3H), 1.67 - 1.49 (m, 2H), 1.45 (d, IH). LCMS-ESr (m/z): !Vi Hj calculated for C21H19CIF2N3O4: 450.10; found: 450.16.
Example 74
Preparation of Compound 74
(lR,4S,12aR)-N-(2-fluoro-3-(triiluoromethyl)berizyl)-7-hydroxy-6,8-dioxo- l^sS^jejSj^J a-octah dro-l^-metlianodi ridotl^-arr^'-dJ razine-^
carboxamide
Figure imgf000147_0002
Compound 74 was prepared in a similar manner to compound 60 using (2-fluoro-3-(trifluoromethyl)phenyl)methanamine in place of (2,3- dich!orophenyl)methanamme. 'H-NMR (400 MHz, DM SO-d6) δ 1 1 .76 (s, I H), 10.48 (t, I H), 8.36 (s, I H), 7.68 (q, 2H), 7.38 (t, I H), 4.68 (s, I H), 4.65 (d, 2H), 4.47 (dd, IH), 3.96 (t, IH), 3.80 (dd, IH), 2.57 (s, IH), 1.88 - 1.69 (m, 3H), 1.67 - 1.50 (m, 2H), 1.45 (d, IH). LCMS-ES ( n/z): [M+H] ; calculated for C22H20F4N3O4: 466.13; found: 466.142.
Example 75
Preparation of Compound 75
(lR,4S^2aR)-N-(3-chloro-4-fluoroben^
octahydro-l ,4-methanodipyrido[l ,2-a: ,2'-d]pyrazine-9-carboxamide
Figure imgf000148_0001
75
Compound 75 was prepared in a similar manner to compound 60 using (3-chioro-4-fiuorophenyi)methanamine in place of (2,3-dichlorophenyl)methanamine. 1 B-NMR (400 MHz, DMSO-d6) δ 1 1.75 (s, I H), 10.43 (t, IH), 8.38 (s, IH), 7.51 (dd, IH), 7.42 - 7.28 (m, 2H), 4.68 (s, IH), 4.51 (d, 2H), 4.47 (dd, IH), 3.97 (t, I H), 3.80 (dd, IH), 2.58 (s, IH), 1.86 - 1.68 (m, 3H), 1.68 - 1.52 (m, 2H), 1 .46 (d, IH). LOIS- ESI+ (m/z): [M+H calculated for C2,H2oCiFN3G4i 432.10; found: 432.159.
Example 76
Preparation of Compound 76
(l R,4S,12aR)-N-((3,5-ditluoropyridin-2-yl)rnethy])-7-hydroxy-6,8-dioxo- \ ,2,3,4,6,8, 12, 12a-octahydro- 1 ,4-methanodipyrido[ 1 ,2-a: 1 ',2'-d]pyrazme-9- carboxamide
Figure imgf000149_0001
Compound 76 was prepared in a simi lar manner to compound 60 using (3,5-difluoropyridin-2-yl)methanamitie in place of (2,3-dichlorophenyl)methanamine. 'H-NMR (400 MHz, Chloroform-d) δ 10.80 (s, 1 H), 8.81 (s, 1H), 8.33 (d. H I ). 7.20 (td, 1H), 4.90 (s, 1H), 4.82 (s, 2H), 4.28 (d, 1H), 3.92 - 3.75 (m, 2H), 3.48 (s, 2H), 1.98 - 1.80 (m, H I. 1.77 (d, 1H), 1.71 ~- 1.58 (m, 2H). LCMS~ESI+ (ni/z) \ M H j calculated for Ο,Π ,.,ΙνΝ ,Ο ,: 417.13; found: 417.189.
Preparation of Compound 77
(lR,4S, 12aR)-7-hydroxy-6,8-dioxo-N-(( )-l -(2,4,6 rifluoroph
1 ,2,3 ,4,6,8, 12, 12a-octahydro- 1 ,4-methanodipyrido[ 1 ,2-a: 1 ',2'-d]pyrazme-9- carboxamide
Figure imgf000149_0002
77 Step 1
A 50-mL round bottom flask was charged with 77- A (0.15 g, 0.39 mmol), (J?)-l-(2,4,6-trifluorophenyl)ethanamine (0.14 g, 0.78 mmol), N,N- diisopropylethylamine (DIPEA) (0.25 g, 1.97 mmol) and HATU (0.29 g, 0.79 mmol) in DCM (10 ml). The reaction mixture was stirred at room temperature for 1 h. The reaction mixture was concentrated down, re-dissolved in EtOAc (50 mL), washed with saturated NaHC03 (2x), saturated NH4CI and dried over Na2S04. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to obtain 77-B as a white solid. LCMS-ESI+ (m/z): [M+H]+ found: 538.
Step 2
A 50-mL round bottom flask was charged with 77-B (0.20 g, 0.37 mmol) in TFA (2 mL). The reaction mixture was stirred at room temperature for 30 min. The solution was concentrated and the residue was purified by flash chromatography using EtOAc-20% MeOH in EtOAc as eluents to afford compound 77. 1H- MR (400 MHz, Chioroform-d) δ 10.67 (d, J = 8.2 Hz, i l l ), 8.22 (s, 1 1 1 ). 6.61 (t, J - 8.4 Hz, 2H), 5.60 (dd, J = 8.1 , 6.9 Hz, 1H), 4.85 (s, 1 H), 3.82 (t, J - 12.2 Hz, ! ! ! }. 3.71 (dd, J = 12.4, 3.4 Hz, 1 H), 2.75 - 2,55 (m, 3H), 1 .97 - 1.57 (m, 9H). 19F-NMR (376 MHz, Chioroform-d) δ -109.65 - -1 1 1 .29 (m), -1 1 1 .76 - -1 13.09 (m). LCMS-ESt (m/z): | \i i ! j found: 448.
Example 78
Preparation of Compound 78
(2R, 13aR)-8-hydroxy-7,9-dioxo-N-((R)- 1 -(2,4,6-trifluorophenyl)ethyl)-
2,3,4,5,7,9, 13, 13a-octahydro-2,5-methanopyrido[ I ',2':4,5]pyrazino[2, 1 - b][l ,3]oxazepine-l 0-carboxamide
Figure imgf000150_0001
78
Figure imgf000151_0001
Figure imgf000151_0002
Step 1
A 50-mL round bottom flask was charged with 78-A (0.30 g, 0.94 mmol), (R)- l-(2,4,6-trifluorophenyl) ethanamine (0.39 g, 1.87 mmol), N,N- diisopropylethyl amine (DIPEA) (0.61 g, 4.87 mmol) and HATU (0.71 g, 1.87 mmol) in DCM (10 ml). The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated down, re-dissolved in EtOAc (50 mL). washed with saturated NaHCOj (2x), saturated NH4CI and dried over Na2S04. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to obtain 78-B as a white solid. LCMS-ES ini/z): [M+Hf ; found: 478.
Step 2
A 50-mL round bottom flask was charged with 78-B (0.4 g, 0.84 mmol) and magnesium bromide (0.4 g, 2.2 mmol) in acetonitrile (5 mL). The reaction mixture was heated to 50 °C. After 10 minutes, the reaction mixture was cooled to 0 °C and ! N hydrochloric acid (4 mL,) was added in. More water (~ 5 mL) was added and the solid was filtrated and washed with water and dried to afford afford compound 78, 1H-NMR (400 MHz, Chloroform-d) δ 12.30 (s, l ! i h 10.59 (d, J = 8.3 Hz, ! H), 8.21 (s, i l l ). 6.60 (t, J = 8.4 Hz, 2H), 5.59 (t, J = 7.4 Hz, 1H), 5.37 (dd, J = 9.4, 4.1 Hz, 1H), 5.31 - 5.09 (m, 1 H), 4.64 (t, J = 3.0 Hz, 1H), 4.20 (dd, J = 12.9, 4.1 Hz, 2H), 3.96 (dd, J = 12.8, 9.4 Hz, 2H), 2.21 - 1.85 (m, 4H), 1.71 - 1.43 (m, 3H). 19F-NM (376 MHz, Chloroform-d) 6 -1 10.37 (tt, J = 8.7, 6.1 Hz), -1 12.19 (t, J = 7.2 Hz). LCMS~ESI+ (m/z): | M H i found: 464. Example 79
Preparation of Compound 79
(1 R,4S, 12aR)-7-hydroxy-6,8-dioxo-N-(2,4,5-trifluorobenzyi)-l,2,3,4,6,8,l 2,12a- octahydro-l,4-methanodipyrido[l ,2-a: ,2'-d]pyrazine-9-carboxamide
Figure imgf000152_0001
Step 1
A 50-mL round bottom flask was charged with 79-A (0.12 g, 0.32 fflffiol), (2,4,5-trifluorophenyl)methanamine (0.10 g, 0.63 mmol), N,N- diisopropy!ethylamine (DIPEA) (0.20 g, 1 .58 mmol) and HATU (0.24 g, 0.63 mmol) in DCM (10 ml). The reaction mixture was stirred at room temperature for 1 h. The reaction mixture was concentrated down, re-dissolved in EtOAc (50 mL), washed with saturated NaHCO-j (2x), saturated NH Cl and dried over Na2SC>4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to obtain 79-B as a white solid. LCMS-ESI+ (m/z): [M+H]+; found: 524.
Step 2
A 50-mL round bottom flask was charged with 79-B (0.15 g, 0.29 mmol) in TFA (2 mL). The reaction mixture was stirred at room temperature for 30 min. The solution was concentrated and the residue was purified by flash chromatography using EtOAc-20% MeOH in EtOAc as eluents to afford compound 79. 'H- MM (400 MHz, Chloroform-d) δ 1 1.70 (s, 1H), 10.65 - 10.18 (m, 1 H), 8.27 (s, 1H), 7.26 (m, 1 H), 6.90 < t l. J = 9.7, 6.4 Hz, 1 H), 4.89 (s, 1 H), 4.60 (d, J = 6.0 Hz, 2H), 4.09 (dd, J = 1 1.4, 2.6 Hz, 1H), 3.96 - 3.66 (m, 2H), 2.68 (s, 1H), 2.15 - 1.43 (m, 6H). i F~NM (376 MHz, Chloroform-d) 8 120.53 - -120.85 (m), -134.68 - -136.79 (m), -142.26 - -144.1 1 (m). LCMS-ESI+ (m/z) [ Vt i i j found: 434.
Example 8Θ
Preparation of Compound 8Θ
(1 R,4S, 12a )-N-(5-chloro-2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo- 1 ,2,3,4,6,8, 12,12a- octahydro-l ,4-methanodipyrido[l ,2-a: r,2'-d]pyrazine-9-carboxamide
Figure imgf000153_0001
80
Step 1
A 50-mL round bottom flask was charged with 80-A (0.12 g, 0.32 mmol), (5-chloro-254-difluorophenyl)methanamme (0.1. 1 g, 0.63 mmol), N,N~ diisopropylethy famine (DIPEA) (0.20 g, 1.58 mmol) and HATU (0.24 g, 0.63 mmol) in EX \1 ( .10 ml). The reaction mixture was stirred at room temperature for 1 h. The reaction mixture was concentrated down, re-dissolved in EtOAc (50 raL), washed with saturated NaHCOj (2x), saturated NH4CI and dried over Na2S04. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to obtain 80-B as a white solid. LCMS-ES ini/z): j VI · ί i | ; found: 541 .
Step 2
A 50-niL round bottom flask was charged with S0~B (0, 14 g, 0.26 mmol) in TFA (2 ml.,). The reaction mixture was stirred at room temperature for 30 minutes. The solution was concentrated and the residue was purified by flash chromatography using Et()Ac-2Q% MeOB in EtOAc as eluents to afford compound 80. Ή-NMR (400 MHz, Chloroform-d) δ 10.46 (s, 1H), 8.27 (s, 1 H), 7.40 (t, J = 7.8 Hz, 1H), 6.89 (t, J = 9.1 Hz, 1H), 4.90 (s, 1H), 4.78 - 4.48 (m, 2H), 4.08 (dd, J = 1 1.3, 2.5 Hz, 1H), 3.95 - 3.63 (m, 2H), 2.68 (s, 1H), 2.22 - 1.51 (m, 7H). 19F-NMR (376 MHz, Chloroform-d) 8 -1 13.37 (q, J = 8.1 Hz), -1 16.37 (q, J = 8.0 Hz). LCMS-E8F (jn/z): [M+H] " found : 451.
Exam le 81
Preparation of Compound 81
(l ,3S,4S,12aS)-3-fluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorohen 1 ,2,3 ,4,6,8, 12, 12a-octahydro- 1 ,4-methanodipyrido[ 1 ,2-a: 1 ',2'-d]pyrazine-9- carboxamide
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000155_0002
Step 1
A 100-mL round bottom flask was charged with 81-A (1.0 g, 3.7 mmol) in DCM (10 mL). The reaction mixture was cooled to 0 UC. Diethylaminosuifur tri fluoride (DAST) (0.58 mL, 4.1 mmol) was slowly added in. Then the reaction mixture was stirred at room temperature for one hour. The mixture was cooled back to 0 °C. Saturated NaHC03 (5 mL) was added dropwise to quench the reaction. Then the reaction mixture was diluted with EtOAc (100 mL), washed with sat. NaHCCh, brine, and dried over Na2S04. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 81-B. LCMS~ESI+ (m/z):
[M+H]+ found: 274. Step 2
A 100-mL round bottom flask was charged with 81~B (0.8 g, 3.0 mmol) in THF (10 mL). The reaction mixture was stirred at -78 °C. 2.0 M LiBH4 in THF (3.2 mL, 6.4 mmol) was slowly added in. Then the reaction mixture was warmed up and stirred at room temperature for 3 hours. Then the reaction mixture was diluted with EtOAc (100 mL) and treated slowly with water (¾ evolution). After the two phases were separated, the aqueous fraction was extracted with EtOAc and the two organic fractions were combined, washed with water, and dried over Na2S04. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 81-C. LCMS~ESI+ (m/z): [M+H]+ found: 246.
Step 3
A 100-mL round bottom flask was charged with 81-C (0.57 g, 2.3 mmol ), triphenylphosphme (1.3 g, 5.1 mmol ) and phthalimide (0.55 g, 3.7 mmol) in THF (15 mL). Then the reaction mixture was cooled to 0 °C with stirring, Diisopropyl azodicarboxvlate (DIAD) (1.0 mL, 5.1 mmol) was slowly added to the reaction mixture. The reaction mixture was stirred at room temperature for overnight. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 81-D. LCMS-ES1+ (m/z): \ \\ i l l found: 375. Step 4
To a solution of 81-D (0.8 g, 2.1 mmol) EtOH (40 mL) was added hydrazine monohydrate (0.6 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate wras concentrated to afford 81-E. LCMS~ESI+ (m/z): [M+H]+ found: 245.
Step 5
A 100-mL round bottom flask was charged with 81-E (0.49 g, 2.0 mmol) and 81-F (0.7 g, 2.0 mmol) in ethanol (7 mL). Sodium bicarbonate (0.34 g, 4.0 mmol) in water (7 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for overnight. The mixture was diluted with EtOAc (50 mL) and washed with water (2 x). The aqueous fractions were extracted with EtOAc (1 x), and the organic fractions were combined, dried ( Na >SO i K and concentrated. The crude 81-G was used for next step without further purification. LCMS-ESI+ (m/z): [M+H]" found: 573.
Step 6
A 100-mL round bottom flask wras charged with 81-G (1 .1 g, 1 .9 mmol) in 4 N HCl /dioxane ( 1 1 mL), Then the reaction mixture was stirred at room temperature for 1 hour. After concentration, 1.0 g intermediate was obtained. The intermediate and DBU (1.3 g, 8.8 mmol) were dissolved in toluene (10 mL). The reaction mixture was heated to 1 10 °C with stirring for 1 hour. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 81-H. LCMS-ES (m/z) [M+H] found: 413.
Step 7
A 100-mL round bottom flask was charged with 81-H (0.56 g, 1 .4 mmol) in THF (5 mL) and MeOH (5 mL). 1 N OH (4 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was acidified by adding 1 N HCl (4 mL), After concentration, the residue was co-evaporated with toluene (3 x). Half of the crude acid, 2,4,6- trifluobenzylamme (0.2 g, 1.3 mmol), N,N-diisopropylethylamine (DIPEA) (0,41 g, 3 , 1 mmol) and HATU (0.48 g, 1 .25 mmol) were dissolved in D F (10 mL). The reaction mixture was stirred at room temperature for 2 hours. The mixture was diluted with EtOAc ( 100 mL) and washed with saturated NaHC03 (2x), saturated M I ι(Ί (2x) and dried over Na2S04. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford 81-1. LCMS-ESI4' (m/z) [M+H] 4 found: 542, Step 8
A 50-mL round bottom flask was charged with 81-1 (0.31 g, 0.58 mmol ) in TFA (3 mL). The reaction mixture was stirred at room temperature for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 81. 1H~NMR (400 MHz, CMoroform-d) δ 10.29 (s, 1H), 8.31 (s, 1H), 6.65 (dd, J = 8.7, 7.5 Hz, 2H), 5.05 - 4.75 (m, 2H), 4.65 (d, J - 5.6 Hz, 2H), 4.11 (d, J = 12.2 Hz, 1H), 3.83 (t, J - 12.3 Hz, lHj, 3.56 (dd, J = 12.3, 3.3 Hz, 1H), 2.77 (s, lH), 2.25 - 1.97 (m, 2H), 1.95 (d, J = 11.0 Hz, 2H), 1.77 id. J = 11.2 Hz, 1 H). 19 F-NMR (376 MHz, Chloroform-d) δ -108.98 (t, J = 8.2 Hz), -1 12.03 (t, J = 7.2 Hz), -168.00. LC S-ESI+ i n/z): found: 452.
Example 82
Preparation of Compound 82
(lS,3R,4R,12aR)-3-fluoro-7-hydroxy-6,8-dioxo-N 2,4,6-trifluorobenzyl)- 1 ,2,3,4,6,8, 12,12a-octahydro- 1 ,4-methanodipyrido[ 1 ,2~a: 1 ',2'-d]pyrazine-9- carboxami.de
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000159_0002
Figure imgf000159_0003
Step 1
A 100-niL round bottom flask was charged with 82- A (0,6 g, 2, 1 mmol) in DCM (6 mL). The reaction mixture was cooled to 0 °C. DAST (0.35 mL, 3.0 mmol) was slowly added in. Then the reaction mixture was stirred at room temperature for one hour. The mixture was cooled back to 0 °C. Saturated NaHCOs (5 mL) was added drop wise to quench the reaction. Then the reaction mixture was diluted with EtOAc (100 mL), washed with sat. aHC03, brine, and dried over Na2S04. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as ements to afford 82-B LCMS-ES1+ (m/z): | M ! ! | found: 274.
Step 2
A ! OO-mL round bottom flask was charged with 82-B (0,4 g, 1.5 mmol) in THF (10 mL). The reaction mixture was stirred at -78 °C. 2.0 M L1BH4 in THF (1.6 mL, 3.2 mmol) was slowly added in. Then the reaction mixture was warmed up and stirred at room temperature for 3 hours. Then the reaction mixture was diluted with EtOAc (100 mL) and added water slowly (¾ evolution). After the two phases were separated, the aqueous fraction was extracted with EtOAc and the two organic fractions were combined, washed with water and dried over Na2S04. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 82-C. LCMS-ESI+ (m/z): | V5 H i found: 246.
Step 3
A 100-mL round bottom flask was charged with 82-C (0.25 g, 1.0 mmol), triphenylphosphme (0.59 g, 2.2 mmol) and phthalimide (0.24 g, 1 .6 mmol) in THF (10 mL). Then the reaction mixture was cooled to 0 UC with stirring. DIAD (0.44 mL, 2.2 mmol ) was slowly added to the reaction mixture. The reaction mixture was stirred at room temperature for overnight. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 82-D. LCMS- ESI+ (m/z): I VI - H i found: 375.
Step 4
To a solution of 82-D (0.35 g, 0.9 mmol) EtOH (20 mL) was added hydrazine monohydrate (0.3 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford 82-E. LCMS-ESI+ (m/z): [M+H]+ found: 245.
Step 5
A 100-mL round bottom flask was charged with 82-E (0.21 g, 0.87 mmol) and 82-F (0.3 g, 0.87 mmol) in ethanol (7 mL). Sodium bicarbonate (0.15 g, 1 .7 mmol) in water (7 ml.) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for overnight. The mixture was diluted with EtOAc (50 mL) and washed with water (2 x). The aqueous fractions were extracted with EtOAc, and the organic fractions were combined, dried (Na2S04), and concentrated. The cnide 82-G was used for next step without further purification, LCMS-ESF (m/z): [M+H] " found: 573.
Step 6
A lGQ-mL round bottom flask was charged with 82-G (0.49 g, 0,86 mmol) in 4 N HCl /dioxane (5 mL). Then the reaction mixture was stirred at room temperature for I hour. After concentration, 0.4 g intermediate was obtained. The intermediate and DBU (0.6 g, 4.0 mmol) were dissolved in toluene (10 mL). The reaction mixture was heated to 1 10 °C with stirring for 1 hour. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 82-H. LCMS-ESF (m/z): [M+H] found: 13.
Step 7
A 1.00-mL round bottom flask was charged with 82-H (0.2 g, 0.49 mmol ) in THF (5 mL) and MeOH (5 mL). 1 N OH (1.5 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for I hour. The reaction mixture was acidified by adding 1 N HCl (1.5 mL). After concentration, the residue was co-evaporated with toluene (3 x). The crude acid, 2,4,6-trifluobenzylamine (0.15 g, 0.95 mmol), N.N-diisopropylethyiamiiie (DIPEA) (0.31 g, 2.4 mmol) and HATU (0.36 g, 0.95 mmol) were dissolved in DCM (10 mL). The reaction mixture was stirred at room temperature for 2 hours. The mixture was diluted with EtOAc (100 mL) and washed with saturated aHC03 (2x), saturated NH4C1 (2x) and dried over Na2S04. After concentration, the crude was purified by column chromatography on silica gei with hexane-EtOAc to afford 82-1. LCMS-ESF (m/z): [M+H]4 found: 542.
Step 8
A 50-mL round bottom flask was charged with 82-1 (0.22 g, 0.41 mmol) in TFA (3 mL). The reaction mixture was stirred at room temperature for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 82, iH~NMR (400 MHz, Chloroform-d) δ
10.25 (s, IH), 8,28 (s, 1H), 6,65 (s, 2H), 5,15 - 4,77 (m, 2H), 4.65 (s, 211), 4.32 - 3.41 (m. 2H), 2.78 (s, 1H), 1.86 (del J = 144.8, 72.3 I ! .. 6H). 19F-NMR (376 MHz, Chloroform-d) δ -108.98 (t, J = 8.2 Hz), -112.03 (t, J = 7.2 Hz), -168.00. LCMS-ESI+ (ni/z): found: 452.
Example 83
Preparation of Compound 83
(18,4R,12a8)-33-difiuoro~7~hydroxy~6,8~dw
1, 2,3,4,6,8, 12 J.2a-octahydro- 1, 4-methanodipyri.do[ 1 ,2-a: ,2'-d]pyrazine-9- carboxamide
Figure imgf000162_0001
8 1,3
Figure imgf000163_0001
Figure imgf000163_0002
Figure imgf000163_0003
Figure imgf000163_0004
S3
Step 1
A 100-mL round bottom flask was charged with 83-A (1.0 g, 3.7 mmol) in DCM (20 mL). The reaction mixture was cooled to 0 °C. Dess-Martin periodinane (1.8 g, 4.2 mmol) was slowly added in. Then the reaction mixture was stirred at room temperature for 3 hours. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 83-B. LCMS~ESI+ (m/z): [M+Hf found: 270. Step 2
A 100-mL round bottom flask was charged with 83-B (0.85 g, 3.2 mmol) in DCM (15 mL). The reaction mixture was cooled to 0 °C. DAST (1.5 mL, 1 1 .3 mmol) was slowly added in. Then the reaction mixture was stirred at room temperature overnight. The mixture was cooled back to 0 °C. Saturated NaHC03 (5 mL) was added dropwise to quench the reaction. Then the reaction mixture was diluted with EtOAc (100 mL), washed with sat. NaHCO¾, brine, and dried over Na2S04. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 83-C. LCMS-ESI+ (m/z): [M+H]+ found: 292.
Step 3
A 100-mL round bottom flask was charged with 83-C (0.44 g, 1.5 mmol) in THE (6 mL). The reaction mixture was stirred at -78 ,JC. 2.0 M LiBH4 in THE (1.6 mL, 3.2 mmol) was slowly added in. Then the reaction mixture was warmed up and stirred at room temperature for 3 hours. Then the reaction mixture was diluted with EtOAc (100 mL) and added water slowly (H2 evolution). After the two phases were separated, the aqueous fraction was extracted with EtOAc and the two organic fractions were combined, washed with water, and dried over a2S04. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 83-D. LCMS-ESI+ (m/z): [M+Hf found: 264.
Step 4
A 100-mL round bottom flask was charged with 83-D (0.17 g, 0.65 mmol), triphenylphosphine (0.37 g, 1.4 mmol) and phtbalimide (0.15 g, 1.0 mmol) in THF (10 mL). Then the reaction mixture was cooled to 0 °C with stirring. DIAD (0,28 mL, 1.4 mmol) was slowly added to the reaction mixture. The reaction mixture was stirred at room temperature for overnight. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 83-E. LCMS- ESI+ (m/z): [M+H found: 393. Step 5
To a solution of 83-E (0.25 g, 0.64 mmol) EtOH (20 ml.) was added hydrazine monohydrate (0.3 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford 83-F. LCMS-ES (m/z): [M+H]+ found: 263.
Step 6
A 100-mL round bottom flask was charged with 83-F (0.18 g, 0.69 mmol) and 83-G (0.324g, 0.69 mmol) in ethanol (7 mL). Sodium bicarbonate (0.12 g, 1.4 mmol ) in water (7 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature overnight. The mixture was di luted with EtOAc (50 mL) and washed with water. The aqueous fractions were extracted with EtOAc, and the organic fractions were combined, dried (Na2S04), and concentrated. The crude 83-H was used for next step without further purification. LCMS-ESr (m/z): [M+H]+ found: 591.
Step 7
A 100-mL round bottom flask was charged with 83-H (0.4 g, 0.68 mmol ) in 4 N HCl /dioxane (3.8 mL). Then the reaction mixture was stirred at room temperature for 1 hour. After concentration, 0.35 g intermediate was obtained. The intermediate and DBU (0.51 g, 3.3 mmol) were dissolved in toluene (10 mL). The reaction mixture was heated to 110 °C with stirring for 1 hour. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 83-1. LCMS-ESf (m/z): [M+H] + found: 431.
Step 8
A 100-mL round bottom flask was charged with 83-Ϊ (0.2 g, 0.47 mmol) in THF (5 mL) and MeOH (5 mL). 1 N KOH (1.4 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was acidified by adding 1 N HCl (1.4 mL). After concentration, the residue was co-evaporated with toluene (3 x). The crude acid, 2,4,6~trifiuobenzylamme (0.14 g, 0.91 mmol), Ν,Ν-diisopropylemylarmne (DIPEA) (0.29 g, 2.2 mmol) and HATU (0.35 g, 0.91 rnmol) were dissolved in DCM (10 mL). The reaction mixture was stirred at room temperature for 2 hours. The mixture was diluted with EtOAc (100 mL) and washed with saturated NaHC03 (2x), saturated NH4CI (2x) and dried over 'Na2S04. After concentration, the crude was purified by column chromatography on si lica gel with hexane-EtOAc to afford 83-J. LCMS-ESI+ (m/z): [M+Hf found: 560.
Step 9
A 50-mL rbf was charged with 83-J (0.1 8 g, 0.32 rnmol) in TFA (3 mL). The reaction mixture was stirred at room temperature for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOe to afford compound 83 as a white solid. Ή-NMR (400 MHz, Chloroformed) δ 10.29 id. J = 6.1 Hz, 1 H), 8.34 (s, 1H), 6.65 (dd, J = 8.7, 7.5 Hz, 2H), 4.83 (s, 1 H), 4.72 - 4.58 (m, 2H), 4.36 - 4.10 (m, 2H), 4.05 (t, J = 1 1.5 Hz, 1H), 2.97 (d, J = 4.4 Hz, 1H), 2.49 - 2.08 (m, 3H), 2.12 - 1.94 (m, 2H). 19F~ MR (376 MHz, Chloroformed) 8 - 92.32 (ddd, J = 225.6, 22.5, 9.1 Hz), -107.64 - -109.54 (m), 12.05 (t, J = 7.0 Hz), -1 14.67 (d, J = 226.7 Hz). LCMS-ESI+ (m/z): found: 470.
Exam le 84
Preparation of Compound 84
(l S,2R,4S,12aR)-7-hydroxy-2-methy[-6,8-dioxo-N-(2,4,6-trifluorobenzy[)- 1 ,2,3 ,4,6,8, 12, 12a-octahydro- 1 ,4-methanodipyrido[ 1 ,2-a: 1 ',2'-d]pyrazme-9- carboxamide
Figure imgf000166_0001
84
Figure imgf000167_0001
S4-E
Hydrazine
Figure imgf000167_0002
Y O OBrs
84-G
Figure imgf000167_0003
84-J
84-I
Figure imgf000167_0004
Step 1
A 100-mL round bottom flask was charged with 84-A (1 ,6 g, 5.9 mmo ) in DCM (20 niL). The reaction mixture was cooled to 0 °C. Dess-Martin periodinane (4.9 g, 11.7 mmol) was slowly added in. Then the reaction mixture was stirred at room temperature for 3 hours. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 84-B. LCMS~ESI+ (m/z): [M+Hf found: 270. Step 2
A 100-mL round bottom flask was charged with 84~B (1.3 g, 4.8 mmol) in THF (30 mL). The reaction mixture was cooled to 0 °C. Tebbe reagent (0.5 M in toluene, 19.4 mL, 9.7 mmol) was slowly added in. Then the reaction mixture was stirred at room temperature for 2 hours. The mixture was cooled back to 0 °C. Saturated NaHC03 (5 mL) was added drop wise to quench the reaction. The reaction mixture was stirred at room temperature for another 15 minutes and filtered through celite. The filtered cake was washed with DCM (2 x). The combined filtrates were concentrated in vacuum and the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford S4~C. LCMS~ESI+ (m/z): [M+H]+ found: 268.
Step 3
To a solution (purged with N2) of 84-C (0.9 g, 3.4 mmol) in EtOH (20 mL) was added Pd/C (0.18 g). The mixture was stirred under H2 for 3 hours. The mixture was filtered through ceiite and the filtrate was concentrated to afford 84-0. LCMS-ES (m/z): j VI · 1 1 1 found: 270.
Step 4
A 100-mL. round bottom flask was charged with 84~D (0.9 g, 3.3 mmol) in THF (6 mL). The reaction mixture was stirred at -78 °C. 2.0 M LiBH4 in THF (13.2 mL, 26.4 mmol ) was slowfy added in. Then the reaction mixture was wanned up and stirred at room temperature for 3 hours. Then the reaction mixture was diluted with EtOAc (100 mL) and added water slowly (H? evolution). After the two phases were separated, the aqueous fraction was extracted with EtOAc and the two organic fractions were combined, washed with water, and dried over Na2S04. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 84- K LCMS-ES1+ (m/z): [M+FI]+ found: 242. Step 5
A 100-mL round bottom flask was charged with 84-E (0.4 g, 1 .66 mmol), triphenylphosphine (0.96 g, 3.6 mmol) and phthalimide (0.39 g, 2.7 mmol) in THF (15 mL). Then the reaction mixture was cooled to 0 l'C with stirring. DIAD (0.7 mL, 3,6 mmol) was slowly added to the reaction mixture. The reaction mixture was stirred at room temperature for overnight. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 84-F. LCMS- KS! (m/z): \ \\ i l l found: 371 .
Step 6
To a solution of 84-F (0.55 g, 1.5 mmol) EtOH (20 mL) was added hydrazine monohydrate (0.3 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford 84-G. LCMS-ESF (m/z): [M+H] found: 241.
Step 7
A 100-mL round bottom flask was charged with 84-G (0.35 g, 1 .4 mmol) and 84-H (0.5g, 1.4 mmol) in ethanol (10 mL). Sodium bicarbonate (0.24 g, 2.8 mmol) in water (10 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for overnight. The mixture was diluted with EtOAc (50 mL) and washed with water (2 x). The aqueous fractions were extracted with EtOAc, and the organic fractions were combined, dried (Na2S04), and concentrated. The crude 84-1 was used for next step without further purification. LCMS-ESF (m/z): [M+H]" found: 583.
Step 8
A 100-mL rbf was charged with 84-Ϊ (0.84 g, 1.4 mmol) in 4 N HC1 /dioxane (8.2 mL). Then the reaction mixture was stirred at room temperature for 1 hour. After concentration, 0.74 g intermediate was obtained. The intermediate and DBU (1.1 g, 7.2 mmol) were dissolved in toluene (10 mL), The reaction mixture was heated to 1 10 tJC with stirring for 1 hour. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 84-J. LCMS-ESI+ (m/z): [M+HJ+ found: 409,
Step 9
A IGO-mL round bottom flask was charged with 84-J (0.4 g, 0.98 mmol) in THF (5 mL) and MeOH (5 mL). 1 N OH (3.0 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was acidified by adding 1 N HCl (3.0 mL), After concentration, the residue was co-evaporated with toluene (3 x). The crude acid, 2,4,6-trifIuobenzylamine (0.32 g, 1.96 mmol), , -diisopropylethy!amine (DI PEA) (0.63 g, 4.9 mmol) and HATU (0.74 g, 1.9 mmol ) were dissolved in DCM (10 mL). The reaction mixture was stirred at room temperature for 2 hours. The mixture was diluted with EtOAc (100 mL) and washed with saturated NaHC0 (2x), saturated NH Cl (2x) and dried over Na2S04. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford 84-K. LCMS~ESI+ (m/z): [M+H]+ found: 538.
Step 10
A 50-mL round bottom flask was charged with 84-K. (0.5 g, 0.93 mmol ) in TFA (6 mL). The reaction mixture was stirred at room temperature for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 84. 3 H-NMR (400 MHz, Chioroform-d) δ 10.37 (s, 1H), 8.28 (s, 1H), 6.65 (t, J = 8.1 Hz, 2H), 4.80 (s, 1H), 4.77 - 4.52 (m, 3H), 4.08 (d, J - 13.1 Hz, 1 1 1 ). 3.88 (d, J = 12,3 Hz, 1 H), 2.47 (d, J = 3.2 Hz, 1 H), 2,35 (s, 1H), 2.16 (ddd, J = 14.3, 1 1.2, 3.6 Hz, 1 H), 1 ,93 - 1 ,57 (m, M l ). 1 ,29 - 1.19 (m, 1 H), 1.1 7 (d, J = 7.0 Hz, 3H). WF-NMR (376 MHz, Chloroform-d) δ -109.24 , -1 1 1 .98. LC MS-ES I ' (m/z): found: 448.
Example 85
Preparation of Compound 85
(6aS,7R, 11S)-1 -hydroxy-2, 13-dioxo-N-(2,4,6-trifluorobenzyl)~6,6a,7,8,9, 10, 1 1 ,13- oct.ahydro-2H-7, l l-m.ethanopyrido[r,2':4,5]pyrazino[l ,2-a]azepine-3-carboxamide
Figure imgf000171_0001
1} H,, Pd/C. HO
Figure imgf000171_0002
85-A 85-B 85-C
Figure imgf000171_0003
85
Step 1
A solution of 85-A (1 100 mg, 3,855 mmol) in DMSO (6 mL) and water (0.75 mL) was stirred at room temperature as N-iodosuccinmide (885 mg, 3,934 mmol) was added. After 2 h, additional N-iodosuccinmide (88 mg, 0.391 mmol) was added and the resulting mixture was stirred at room temperature for 1.5 h. The dark brown reaction mixture was diluted with EtOAc, and washed with a mixture of 10 % aq. a2S20a solution and aq. NaHC03 solution (~1 :4 mixture) and then with water (with some brine). After the aqueous fractions were extracted wit EtOAc, the organic fractions were combined, dried ( a2S04), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to obtain 85-B. 3'H-NM (400 MHz, CDC13) δ 7.51 - 7.44 (m, 2H), 7.33 - 7. 17 (m, M i l 4.22 - 4.05 (m. 2H), 4.02 - 3.86 (m, 21 1 ). 3.77 (d, J = 5.3 Hz, 1H), 3.54 - 3.44 (m, 1H), 3.27 (t, J = 4.5 Hz, 1H), 2.75 - 2.66 (m, 1H), 2.30 (dddd, J = 14,8, 13.1 , 7.2, 5.8 Hz, 1H), 2.14 (dddd, J = 14.8, 13.0, 6.1 , 2.1 Hz, i l l ), 1.97 (d, J = 8.9 Hz, 1H), 1.58 - 1.46 (m, 1 H), 1.45 - 1.34 (m, 4H), 1 .24 (t, J - 7.1 Hz, 3H). LCMS-ES (m/z): [M+H]'1' calculated for C : , I !■ XO - 430.1 ; found: 430.0.
Step 2
A solution of 85-B (993 mg, 2.313 mmol), AIBN (305 mg, 1.857 mmol), and tributyltin hydride (1392 mg, 4,799 mmol) in toluene (15 mL) was stirred at 100 °C. After 2 h, the reaction mixture was cooled to room temperature, diluted with EtOAc, and washed with water and brine. After the aqueous fractions were extracted with EtOAc, the organic fractions were combined, dried (Na2S04), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to obtain 85-C. IH-NMR (400 MHz, CDC! 3) δ 7.57 - 7.49 (m, 2H), 7.32 - 7.23 (m, 2H), 7.23 - 7.15 (m, 1H), 4.24 - 4.02 (m, 2H), 3.97 (q, J = 6.7 Hz, 1H), 3.83 (d, J = 5.1 Hz, 1 H), 3.48 (t, J = 4.6 Hz, 1H), 3. 19 - 3.04 (m, 1H), 2.58 (p. J = 4.0 Hz, 1H), 2.30 (dddd, J = 14.7, 13.1 , 7.0, 4.5 Hz, 1H), 1.98 (d, J = 1 1.2 Hz, 1H), 1.64 (tdd, J = 13.3, 6.2, 2.6 Hz, 1 H), 1 .49 - 1.33 (m, 31 1 ). 1.37 (d, J = 6.7 Hz, 3H), 1.32 - 1.26 (m, 1H), 1.23 (t, J = 7.2 Hz, 3H). LCMS-ES (m/z) [M+H]+ calculated for <YJ i„\0 : . 304.2; found: 304.1.
Step 3
A mixture of 85-C (725 mg, 2.39 mmol) and 20% Pd(OH)2/C (35 1 mg) in EtOH (25 mL) and 4 N HCl in dioxane (0.9 mL) was stirred under H2 atmosphere. After 2 h, the reaction mixture was filtered, and the filtrate was concentrated. LCMS- ESI+ (m/z): [M+H calculated for CioHi8N03: 200.13; found: 200.1 . After the residue was co-evaporated with toluene (x 2), the residue and Boc20 (720 mg, 3.299 mmol) in THF (15 mL) was stirred at room temperature as Ν,Ν-diisopropyiethylamine (DIPEA) (1.2 mL, 6,889 mmol) was added. After 1 h, the reaction mixture was diluted with water and extracted with EtOAc (x 2). After the organic extracts were washed with water, the combined extracts were dried (Na2S04) and concentrated. The residue was purified by flash using hexanes - EtOAc as eluents to obtain 85-D which appears to be a mixture of retainers. 1H- M (400 MHz, CDC13) δ 4.42 - 3.97 (m, 5H), 2.62 (d, J = 5.6 Hz, 1H), 2.45 - 2.26 (m, 1H), 2.25 - 2.15 (m, 1H), 1.80 (td, J = 13.7, 6,7 Hz, 1H), 1.66 (dd, J = 12.3, 6.6 Hz, 2H), 1.55 - 1.70 (rn, 2H), 1.47 (s, 2H), 1 .42 (s, 7H), 1.28 (dt, J = 9,5, 7.1 Hz, 3H). LCMS-ESI+ (m/z): [ \1 I I I calculated for ( ,ΝΟ^ 300.2; found: 299.7.
Step 4
To a solution of 85-D (568 mg, 1.897 mmol) and pyridine (0.25 mL, 3.091 mmol) in THF (5 mL) was added phenyl chlorothionoformate (0.3 mL, 2.169 mmol) at 0 °C, which produced insoluble material quickly. After -30 min at 0 °C, additional pyridine (0,3 mL, 3.709 mmol) and phenyl chlorothionoformate (0.3 mL, 2.169 mmol) were added. After 1 .5 h at 0 °C and 1 h at room temperature, the mixture was concentrated, and the residue was dissolved in EtOAc and water. After separation of two layers, the organic fraction was washed with -0.1 N HC1, saturated aqueous NaHC03, and brine . After the aqueous fractions were extracted with EtOAc, the combined organic fractions were dried (Na2S04), and concentrated. The residue was purified by flash chromatography using EtOAc/hexanes as eluents to afford 85-E. H- NMR (400 MHz, CDC13) δ 7,47 - 7.37 (m, 2H), 7,30 (t, J = 6,9 Hz, 1 1 1 ), 7.1 1 (dd, J = 8.0, 4.0 Hz, 2H), 5.54 (dt, J = 9.0, 4.9 Hz, 1 1 1 ), 4.50 (dt, J - 9.8, 5.3 Hz, I H), 4.35 (dd, J = 21 .4, 5.0 Hz, 1 H), 4.30 - 4.14 (m, 21 1 ). 2.71 (s, 1 H), 2.54 (s, 1 H), 2.14 - 2.00 (m, 1 H ), 1.82 (m, 31 1 ). 1 .54 (m, 1 1 1 ). 1.48 (s, 4. H), 1.45 (s, 4.5 ! I i. 1 .30 (dt, J = 9.4, 7.1 Hz, M l ). LCMS-ESI+ (m/z): j \1 Π ] calculated for ί A \ ,SO,S : 436.2; found: 435.8.
Step 5
A mixture of 85-E (602 mg, 1.382 mmol), ΑΓΒΝ (182 mg, 1.108 mmol), and tributyltin hydride (608 mg, 2.096 mmol) in toluene (8 mL) was stirred at 100 °C. After 1 h, the reaction mixture was concentrated and the residue was dissolved in EtOAc before washing with water and brine. After the aqueous fractions were extracted with EtOAc, the combined organic fractions were dried (Na2S0 ) and concentrated. The residue was purified with flash chromatography using EtOAc/hexanes as e!uents to give 85-F which appears to be a mixture of rotamers. *H- NMR (400 MHz, CDC13) δ 4.37 - 4.06 (m, 4H), 2.69 - 2.53 (m, 1H), 2.1 1 (m, I H), 1 .97 (m,0.65H), 1.93 - 1.80 C m. 1.35H), 1.54 (s, 5H), 1.46 (s, 3.15H), 1.42 (s, 5.85H), 1.27 (m, 3H). LCMS~E8I+ (ni/z): [M-C4HS+H]+ calculated for CiiHi 8N04: 228.1 ; found: 227.9. Step 6
85-F (420 mg) was repurified and the purified 85-F in THF (3 mL) was stirred at 0 °C as 2.0 M LiBH4 in THF (1.5 mL) was added. After 5 min, the mixture was stirred at room temperature for 17 h and additional 2.0 M L1BH4 in THF (1 .5 mL) was added at room temperature. After 23 h at room temperature, additional 2.0 M L1BH4 in THF (3 mL) was added and the resulting mixture was stirred for -72 h. After the reaction mixture was stirred at 0 °C as water was slowly added and further diluted with water, the product was extracted with EtOAc (x 2). The extracts were washed with water, combined, dried (Na2S0 ), and concentrated. The residue was purified by flash chromatography using hexane - EtOAc as e!uents to give S5-G, 1H-N R (400 MHz, CDCI3) δ 4.12 (t, J = 5.3 Hz, 1H), 3.99 (dd, J = 12.0, 7.9 Hz, IH), 3.85 (dd, J = 8.0, 4.7 Hz, I H), 3.73 (dd, J = 1 1.9, 1.4 Hz, IH), 2.28 (d, J = 4.6 Hz, I H), 1 .90 - 1.73 (m, 2H), 1.68 - 1.45 (m, 6H), 1.47 (s, 9H), 1.43 - 1.33 (m, IH). LCMS-ESI+ (m/z): \ V ·· C4H8+H]";" calculated for C9H16NQ3: 186.1 ; found: 186.0. Step 7
A solution of 85-G (1 98 mg, 0.820 mmol), phthalimide (200 mg, 1.359 mmol), and PPh3 (488 mg, 1.861 mmol) in THF (10 mL) was stirred at 0 °C bath as DIAL) (0.36 mL, 1.828 mmol) was added. After 30 min at 0 °C, the mixture was stirred at room temperature for 17 h. The reaction mixture was concentrated and the residue was purified by flash chromatography using hexane-EtOAc as eiuents to 85-H which appears to be a mixture of rotamers. 1H- MR (400 MHz, CDCI3) δ 7.82 (dd, J = 5.4, 3.1 Hz, 2H), 7.69 (dd, J = 5.4, 3.1 Hz, 2H), 4.46 (s, IH), 4.19 (m, 2H), 3.95 (s, IH), 2.31 - 2.14 (m, 1 H), 2.05 id. J = 16.5 Hz, lH), 1.84 (m, 2H), 1 .79 - 1.70 (m, i l l ), 1.66 (m, 1H), 1.61 - 1.30 (m, 12H). LCMS-ES (rn/z): [M +H] calculated for C21H27N204: 371.2; found: 370.8. Step 8
To a solution of 85-H (270 mg, 0.729 mmol) in EtOH (12 mL) was added hydrazine hydrate (0.145 mL, 3.083 mmol) at room temperature and the resulting solution was stirred at 70 °C. After 1.5 h, the mixture was cooled to 0 °C and diluted with ether (30 mL) before stirring for 1 h at 0 °C. The mixture was filtered and the filtrate was concentrated. The residue was dissolved in CH2Ci2 and filtered to remove some insoluble material . The resulting filtrate was concentrated. The residue, combined with 85-Ϊ (257 mg, 0.742 mmol), and NaHC03 (131 mg, 1.559 mmol) in water (3 mL) and EtOH (3 mL) was stirred at room temperature. After 1 h, the mixture was diluted with water and extracted with EtOAc (x 2). After the extracts were washed with water, the organic extracts were combined, dried (^¼804), and concentrated. To a solution of the residue in CH2C3.2 (2 mL) was added 4 N HCl in dioxane (6 mL). After 1 .5 h at room temperature, the solution was concentrated and co-evaporated with toluene, A mixture of the residue and DBU (0.6 mL, 4,012 mmol) in toluene (5 mL) was stirred at 100 °C bath. After 1 h, additional DBU (0.3 mL, 2.006 mmol) was added and the mixture was stirred another 1 h at 100 "C. After the mixture was concentrated, the residue was purified by flash chromatography using EtOAc - 20% MeOH/EtOAc as eiuents to give S5~J. ¾-NM (400 MHz, CDC13) 6 8.08 (s, 1H), 7.71 - 7.62 (m, 2H), 7.36 - 7.29 (m, 2H), 7.29 - 7.23 (m, i l l ), 5.44 id. J = 9.8 Hz, 1 H), 5.10 (d, J - 9.8 Hz, i l l ), 4.44 - 4.28 (m, 3H), 4.23 (t, J - 13.0 Hz, I I I ). 3.99 (ddt, J = 10.2, 6.3, 3.6 Hz, 2H), 2.44 - 2.36 (m, 1 1 1 ). 2.29 (dt, J - 1 1.6, 5.3 Hz, H I }. 1 .84 (dt, J i O.K. 5.3 Hz, 2H), 1 .77 - 1 .61 (m, 3H), 1.57 (d, J = 1 1 .7 Hz, 1 H), 1 .48 (ddd, J - 20.9, 12.3, 5.5 Hz, 1H), 1.38 (t, J = 7.1 Hz, 31 1 ). f X WiS-ESf (m/z): [M +H calculated for ! > ·Χ ·.{),: 423.2; found: 423.3. Step 9
A mixture of 85- J (214 mg, 0.507 mmol) in THF (4 mL) and MeOH (4 mL) was stirred at room temperature as 1 N KOH (1 .1 mL) was added. After 30 min, the reaction mixture was concentrated to ~1 mL, acidified with 1 N HQ (~1.2 raL), and diluted with brine before extraction with CH2Ci2 (20 mL x 2). The combined extracts were dried (Na2S04) and concentrated to obtain the crude acid. LCMS-ESI+ (m/z): [M +H]+ calculated for C22H23N2O5 : 395.2; found: 395.3.
A mixture of the crude acid (199 mg, 0.505 mmol), 2,4,6-trifluorobenzyl amine (130 mg, 0.807 mmol), and HATU (304 mg, 0.800 mmol) in CH2Ci2 (6 mL) was stirred at room temperature as N.N-diisopropylethyiamine (DIPEA) (0.62 mL, 3.559 mmol) was added. After 30 mm, the reaction mixture was concentrated and the residue was dissolved in EtOAc, washed with saturated aqueous NH4C1 (x 2), saturated aqueous NaHC03 (x 2), and brine. After the aqueous fractions were extracted with EtOAc, two organic fractions were combined, dried (Na2S04) and concentrated. The residue was purified by flash using EtOAc-20%MeOH/EA as eluents to obtain 85- . Ή-NMR (400 MHz, CDCI3) δ 10.40 (t, J = 5.7 Hz, IH), 8.42 (s, 1 H), 7.68 - 7.54 (m, 2H), 7.33 (ddd, J = 7.7, 6.3, 1.5 Hz, 2H), 7.30 - 7.26 (m, IH), 6.74 - 6.60 (m, 2H), 5.37 (d, J = 10.0 Hz, IH), 5.17 (d, J = 10.0 Hz, IH), 4.76 - 4.57 (m, 2H), 4.46 (dd, J = 6.0, 4.3 Hz, I H), 4.34 (t, J = 12.4 Hz, IH), 4.07 (dd, J = 12.4, 3.6 Hz, I H), 3.91 (dt, J = 12.4, 3.9 Hz, I H), 2.52 - 2.44 (m, I H), 2.32 (dd, J = 1 1 .8, 6.2 Hz, IH), 1 .92 (dt, J = 10.7, 5.4 Hz, I H), 1 .83 - 1.70 (m, 3 H i. 1 .67 (d, J = 1 1 .7 Hz, I H), 1 .52 (dddt, J - 25.5, 17.0, 1 1 .8, 5.3 Hz, 2H). "F-NMR (376 MHz, CDCI3) δ -109.15 (dq, J ==: 15.0, 7.5, 7.1 Hz, I F), - 1 1 1.85 (t, J = 6.8 Hz, 2F). LCMS-ES (m/z): [M +Hf calculated for C29H27F3N3O4: 538.2; found: 538.3.
Step 10
85-K (1 87 mg, 0.348 mmol) was dissolved in trifluoroacetic acid (3 mL) at room temperature and stirred at room temperature. After 1 h, the solution was concentrated and the residue was dissolved in CH2CI2. After the solution was washed with 0.1 N HC1, the aqueous fraction was extracted with CH2C12 (x 2). The organic fractions were combined, dried ( \';s -SO . }, and concentrated. The residue was purified by flash chromatography using CH2Cl2-20% MeOH in ( I LCL as eluents to obtain 150 mg (96%) of compound 85. Compound 85 was further purified by recrystallization from methanol (10 mL) to give compound 85. ' l l-N M R (400 MHz, CDCI3) δ 12.09 (s, I H), 10.39 (t, J - 5.7 Hz, I H), 8.36 (s, IH), 6.74 - 6.48 (m, 2H), 4.64 (d, J = 5.7 Hz, 2H), 4.59 (dd, J = 6.1, 4.4 Hz, 1H), 4.36 - 4.18 (m, 2H), 4.12 (dt, J = 12.4, 4.1 Hz, 1H), 2.68 - 2.47 (m, 1H), 2.25 - 2.10 (m, III .2.10 - 1.98 (m, 111).1.98 - 1.66 (m, 4H), 1.66 - 1.48 (ra, 2H). "F-NMR (376 MHz, CDC13) δ -109.23 (ddd, J = 15.1, 8.6, 6.0 Hz, IF), - 112.02 (t, J = 6.9 Hz, 2F). LCMS-ESf (m/z) [M +H]+ calculated for C22H21F3 3O4: 448.2; found: 448.3.
Example 86
Preparation of Compound 86
(1 R,3S,4R, 12aS)-7-hydroxy-3-m.ethyi-6,8-dioxo-N-(254,6-trif].uorobenzyi)- 1, 23,4,6,8, 12 J.2a-octahydro- 1, 4-methanodipyri.do[ 1 ,2-a: ,2'-d]pyrazine-9- carboxamide
Figure imgf000177_0001
e
Figure imgf000178_0001
86-1
1 ) Η,ΝΝΗ,
Figure imgf000178_0002
Figure imgf000178_0003
Step 1
A solution of 86-A (10,160 g, 39.48 mmol) in DMSO (52 ml,) and water (6.5 ml,) was stirred at room temperature as N-iodosuccinmide (8.888 g, 39.50 mmol) was added. After 30 min, the dark brown reaction mixture wras diluted with EtOAc, and washed with saturated aqueous 'NaHC0 solution, 10 % aqueous a2S203 solution ],and brine. After the aqueous fractions were extracted with EtOAc, the organic fractions were combined, dried (Na2SC>4), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to obtain 86~B as a white solid. 1H~ NMR (400 MHz, CDCi,) 8 7.33 - 7.19 (m, 5H), 4.25 - 4.12 (m, 1H), 3.79 (q, J = 1.6 Hz, 1 H), 3.72 (q, J = 6.5 Hz, IH), 3.51 (s, H i ). 3.47 (s, 3H), 3.31 (dd, J - 3.9, 1.6 Hz, I H), 2.76 - 2.69 (m, IH), 2.13 (ddd, J - 14.3, 7.8, 1.7 Hz, H i ). 2.08 - 1.97 (m, I H), 1.91 (did, J - 14.1 , 4.0, 1.5 Hz, I I I . 1.42 (d, J = 6.5 Hz, M l ). LCMS-ES1+ (m/z): [M+H] calculated for ('„.! h . I NO *: 402.1 ; found: 402.0.
Step 2
A solution of 86-B (12.468 g, 31.07 mmol), azobisisobutyronitrile
(AIBN) (4.082 g, 24.86 mmol), and tributyltin hydride (18.047 g, 62.22 mmol) in toluene (150 mL) was stirred at 100 °C. After 30 min, the reaction mixture was cooled to room temperature, diluted with EtOAc, and washed with water and brine. After the aqueous fractions were extracted with EtOAc, the organic fractions were combined, dried (Na2S04), and concentrated. The residue was purified by flash chromatography twice using hexanes - EtOAc as eluents to obtain 86-C. 'H- M R (400 MHz, CDCI3) 5 7.39 - 7.31 (m, 2H), 7.31 - 7.24 (m, 2H), 7.24 - 7.17 (m, 1H), 4.1 1 (s, 1 H), 3.72 (s, I H), 3.49 (s, 3H), 3.33 (d, J = 3.4 Hz, 1H), 3.27 (d, J = 6.4 Hz, I H), 2.65 - 2.51 (m, 1H), 1.92 (ddd, J = 13.6, 6.8, 2.4 Hz, 1H), 1.69 - 1.50 (m, 2H), 1.47 (d, J = 10.1 Hz, IH), 1.41 (d, J 6.6 Hz, 3H), 1.21 - 1.07 (m, IH). LCMS-ESI+ (m/z): | M H j calculated for C16H22 O : 276.2; found: 276.1 .
Step__3
A mixture of 86-C (4.1 87 g, 15.21 mmol ) and 20% Pd(OH)2/C (1 .022 g) in EtOH (100 mL) and 4 N HC1 in dioxane (5.7 mL) was stirred under H2 atmosphere. After 1.5 h, the reaction mixture was filtered, and the filtrate was concentrated. After the residue was co evaporated with toluene, the residue was used for the next step. LCMS-ES (m/z) [M+H]'4 calculated for (' ,! \ > \\0 -. : 172.1 ; found: 172.1.
After the residue was co-evaporated with toluene, the residue and Boc20 (5.712 g, 26.17 mmol) in THF (45 mL) was stirred at room temperature as N,N- diisopropylethyl amine (DIPEA) (8 mL, 45.93 mmol) was added. After 30 min, the reaction mixture was diluted with water and extracted with EtOAc (x 2). After the organic extracts were washed with water, the combined extracts were dried (Na2S04) and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to obtain 86-D, Ή NMR spectrum suggests a mixture of retainers. 1H-NMR (400 MHz, CDC13) δ 4.20 (d, J = 7.6 Hz, IH), 4.19 - 4.10 (m, 2H), 4.08 (d, J = 3.5 Hz, IH), 3.72 (s, 3H), 2.74 (d, J - 5.6 Hz, IH), 1.97 (ddd, J = 13.6, 6.9, 2.8 Hz, ill), 1.88 - 1.78 (m, III . 1.79 - 1.50 (m, 1H), 1.46 (s, 311).1.38 (s, 6H), 1.31 (d, J - 13.3 Hz, 1H). LCMS-ESI+ (m/z): |VI-iii calculated for C1 H22NO5: 272.2; found: 271.6. Step 4
A solution of S6~D (1659 mg, 6.115 mmol) in C'i !.·( ;.· (35 mL) was stirred at 0 °C bath as Dess-Martin periodinane (5.183 g, 12.22 mmol) was added in portions. After 5 rain, the mixture was stirred at room temperature. After 2 h, the reaction mixture was cooled in an ice bath, quenched with water, and filtered. The filtrate was washed with saturated NaHCOs, dried (Na2S04), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to give 86-E. 3H NMR spectrum suggests two rotamers. 1H-NMR (400 MHz, CDCI3) δ 4.43 (d, J = 3.8 Hz, 0.5H), 4.39 (s, 1H), 4.26 (s, 0.5H), 3.75 (s, 3H), 3.10 (s, 1H), 2.24 (d, J = 4.5 Hz, 0.5H), 2.19 (d, J = 4.4 Hz, 0.5H), 2.12 (d, J = 4.4 Hz, 0.5H), 2.07 (d, J = 4.2 Hz, 0.5H), 2.01 (dd, J = 4.5, 2.2 Hz, 0.5H), 1.98 (dt, J = 4.3, 1.9 Hz, 0.5H), 1.80 (s, 0.5H), 1.77 (s, 0.5H), 1.46 (s, 4,51!), 1.40 (d, J = 2.8 Hz, 4.511). LCMS-ESI+ (m/z): [M- C4H8+H] + calculated for ('·,!! ι.Λί),· 214.1; found: 213.8.
Step 5
A solution of 86-E (528 mg, 1.961 mmol) in THF (12 mL) was stirred at
0 °C as 0.5 M solution of Tebbe reagent in toluene (7.9 mL, 3.95 mmol) was added dropwise. After addition, the brown solution was allowed to warm to room temperature slowly and was stirred at room temperature for 2.5 h. The reaction mixture was stirred at 0 °C bath as the reaction was quenched carefully by the addition of saturated aqueous NaHC03 solution. After the mixture was diluted with CH2Cl2 and stirred at room temperature for 15 minutes, the resulting mixture was filtered through celite pad and the filter cake was washed with CH?C1?. After the two fractions in the filtrate were separated, the aq. fraction was extracted with CH2C12, and the organic fractions were combined, dried (Na2SQ4), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to give 86~F. Ή NMR spectrum suggests two rotamers. 1H~ MR (400 MHz, CDC13) δ 5.13 (s, 0.6H), 5.04 (s, 0,411). 4.82 - 4.71 (m, ill).4.55 (s, 0.61!).4.43 (s, 0,411), 4.29 (d, J = 3.7 Hz, 0.411).4.24 (d, J = 3,7 Hz, 0.6! I).3,71 (s, 3H), 2.84 (s, IH), 2.14 (m, 2H), 1.75 (s, 0.6H), 1.74 - 1.70 (s, 0,41!), 1.55 (m, 1H), 1.45 (s, 3.6H), 1.37 (s, 5.4H). LCMS-ESI+ (m/z): [M III calculated for C14H22N04: 268.2; found: 267.6. Step 6
A mixture of 86~F (333 mg, 1.246 mmol) and 20% Pd(OH)2/C (53 mg) in EtOH (5 mL) was stirred under H2 atmosphere. After 30 min, the mixture was filtered and the filtrate was concentrated to give 86-G. lH NMR. spectrum suggests two retainers. 1H-N (400 MHz, CDC13) δ 4.20 (m, 1H), 4.08 (m, 1H), 3.71 (two s, 311). 2.68 (m, IH), 2.06 (m, IH), 1.80 - 1.63 (m, 2H), 1.63 - 1.51 (m, 1H), 1.44 (s, 4H), 1.38 (s, 5H), 1.13 (m, 3H), 0.92 (m, 1H). LCMS-ESI+ (m/z): [M +H]+ calculated for Ci4H2,¾N04: 270.2; found: 269.7.
Step?
A solution of 86-G (336 mg, 1.482 mmol) in THF (5 mL) was stirred at
0 °C as 2.0 M L1BH4 in THF (1.5 mL) was added. After 5 min, the mixture was stirred at room temperature. After 2 h, additional 2.0 M L1BH4 in THF (1.5 mL) was added. After 21 h at room temperature, additional 2.0 M LiBH4 in THF (3 mL) was added. After 3 h at room temperature, the solution was heated at 35 °C for 18 h. The reaction mixture was cooled to 0 °C and quenched carefully with water. After the mixture was extracted with EtOAc (x 2), the two organic fractions were washed with water, combined, dried (Na2S04), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc to give 86-H. Ή-NMR (400 MHz, CDCI3) δ 4.95 - 4.09 (br, !!!}.4.05 (s, IH), 3.82 (dd, J = 11.5, 7.7 Hz, IFF), 3.76 - 3.69 (m, IFF), 3.66 (d, J = 11.5 Hz, IH), 2.45 id..1 = 4.1 Hz, IH), 2,03 (dqdd, J = 11.4, 7.0, 4.5, 2.6 Hz, IH), 1.77 - 1.57 (m, 2H), 1.48 (dd, J = 10.1, 1.8 Hz, IH), 1.45 is.9H), 1.00 (d, J 6. Hz, 3H), 0.93 (ddd, J = 13.2, 4.7, 2.6 Hz, IH). LCMS-ESI+ (m/z): [M ·Ι1| calculated for C13H24NO3: 242.2; found: 241.7. Step 8
A solution of 86-H (218 mg, 0.903 mmol), phthalimide (218 mg, 1.482 mmol), and PPI13 (535 mg, 2.040 mmol) in THF (10 mL) was stirred at 0 °C bath as DIAD (0,40 mL, 2.032 mrnoi) was added. After 10 min at 0 °C, the mixture was stirred at room temperature for 19 h. The reaction mixture was concentrated and the residue was purified by flash chromatography using hexane-EtOAc as eluents to give 86-ϊ . *H NMR suggests two rotamers. ^-N (400 MHz, CDC13) δ 7.82 (dt, J = 7.3, 3.6 Hz, 2H), 7.70 (d, J = 5.3 Hz, 2H), 4.53 - 4.26 (m, !H), 4.26 - 3.89 (m, 2H), 3.89 - 3.65 (m, 1H), 2.28 (m, IH), 2.04 (m, IH), 1.82 - 1.65 (m, 2H), 1.66 - 1.43 (m, 7H), 1.38 (s, 4H), 1.19 - 1.01 (m, 3H). LCMS-ESf (m/z): [M +H]+ calculated for ( , ; ! ί.- Χ -Ο ,: 371.2; found: 370.8. Step 9
To a solution of 86-Ϊ (31 9 mg, 0.861 mrnoi) in EtOH (12 mL) was added hydrazine hydrate (0.17 mL, 3.494 mmol) at room temperature and the resulting solution was stirred at 70 °C bath. After 1.5 h, the mixture was cooled to 0 °C and diluted with ether (25 mL) before stirring for 1 h at 0 °C. The mixture was filtered and the filtrate was concentrated. The residue was dissolved in CH2C12 and filtered to remove some insoluble material. The resulting filtrate was concentrated to give crude amine. LC S-ESI+ (m/z): [M +Hj+ calculated for C13H25N2O2: 241.2; found: 240.9.
After the crude amine was co-evaporated with toluene, a mixture of the crude amine, 85-1 (300 mg, 0.866 mmol), and aHC03 ( 150 mg, 1 .845 mmol ) in wrater (3 mL) and EtOH (3 mL) was stirred at room temperature. After 2 h, the mixture was diluted with water and extracted with EtOAc (x 2). After the extracts were washed with water, the organic extracts were combined, dried (Na2S04), and concentrated. To a solution of the residue in CH2C12 (2 mL) was added 4 N HCl in dioxane (6 mL). After 1.5 h at room temperature, the solution was concentrated and co-evaporated with toluene. A mixture of the residue and DBU (0.65 mL, 4.347 mmol) in toluene (6 mL) was stirred at 100 °C. After 1 h, additional DBU (0.65 mL, 4.347 mmol) was added and the mixture was stirred at 100 °C. Additional DBU (0.65 mL, 4.347 mmol) was added after 1 h and the mixture was stirred another 2.5 h at 100 °C. The mixture was diluted with CH- L and washed with water containing 3 mL of 1 N HCl. The organic fraction was dried (Na2S04) and concentrated. The residue was purified by flash chromatography using EtOAc-20% MeOH/EtOAc as eluents to give 86-J. 1H-NMR (400 MHz, CDCI3) δ 8.09 (s, I H), 7.70 - 7.62 (m, 2H), 7.37 - 7.27 (m, 3H), 5.48 (d, J 9.9 Hz, IH), 5.16 (d, J = 9.9 Hz, IH), 4.53 (s, III).4.38 (m, 2H), 4.11 (m, 1H), 3.97 (dd, J = 12.2, 3.0 Hz, III).3.88 (dt, J - 12.2, 3.0 Hz, 111).2.63 (d, J = 4.2 Hz, III).2.28 (qd, J = 7.2, 3.1 Hz, IH), 2.00 - 1.88 (m, ill).1.80 - 1.56 (m, 2H), 1.39 (t, J = 7.1 Hz, 3H), 1.07 (d, J = 6.9 Hz, 3H), 1.04 (dd, J = 5.0, 2.5 Hz, 1H). LC S-ESI+ (m/z): [M +H]+ calculated for C24H27N2O5 : 423.2; found: 423.2.
Step 10
A mixture of 86- J (83 rag, 0.196 mmol) in THF (2 mL) and EtOH (2 mL) was stirred at room temperature as 1 N OH (0.4 mL) was added. After 30 mitt, the reaction mixture was diluted with water and washed with CH2Cl2. After the aqueous fraction was acidified with 1 N HCI 0,45 mL), the product was extracted with CI !··('!·· (x 2). The combined extracts were dried (Na2S04) and concentrated to obtain the crude acid. LCMS-ESF (m/z): [M +H]+ calculated for C22H23N2O5: 395.2; found: 395.2.
A mixture of the crude acid (69 mg, 0.175 mmol), 2,4,6-trifluorobenzyl amine (42 mg, 0.261 mmol), and HA.TU (106 mg, 0.279 mmol) in CH2Ci2 (3 ml.) was stirred at room temperature as Ν,Ν-diisopropyiethylamine (DIPEA) (0.25 mL, 1.435 mmol ) was added. After 30 min, the reaction mixture was concentrated and the residue was dissolved in EtOAc, washed with saturated aqueous NH4CI (x 2), saturated aqueous NaHC03 (x 2), and brine. After the aqueous fractions were extracted with EtOAc, two organic fractions were combined, dried (Na2S04) and concentrated. The residue was purified by flash chromatography using EtOAc-20%MeOH/EtOAc as eluents to obtain 86-K. 1H- MR (400 MHz, CDC1) δ 10.40 (t, J - 5.7 Hz, IH), 8.40 (s, IH), 7.66 - 7.51 (m, 2H), 7.36 - 7.29 (m, 2H), 7.29 - 7.23 (m, IH), 6.71 - 6.61 (m, 2H), 5.36 (d, J = 10.0 Hz, IH), 5.18 (d, J = 10.0 Hz, IH), 4.73 - 4.58 (m, 2H), 4.53 (s, IH), 4.22 - 4.11 (m, IH), 4.03 (dd, J = 12.4, 3.1 Hz, IH), 3.81 (dt, J = 12.3, 3.1 Hz, IH), 2.68 - 2.59 (m, IH), 2.29 (dddd, J = 11.4, 7.1, 4.7, 2.4 Hz, IH), 1.94 (ddd, J = 13.5, 11.2, 4.6 Hz, IH), 1.88 - 1.67 (m, 2H), 1.06 (d, J = 7.0 Hz, 3H), 1.03-1.09 (m, IH). i9F- MR (376 MHz, CDCI3) 8 -109.14 (ddd, J = 15.2, 8.7, 6.2 Hz, IF), -111.86 (t, J = 7.0 Hz, 2F). LCMS-ESf (mz): [M H| calculated for Ο,Ι -.Λ,Ο,: 538.2; found: 538.1.
Step 11 86- (61 mg, 0, 1 13 mmol) was dissolved in trifluoroacetic acid (2 mL) and stirred at room temperature. After 1 h, the solution was concentrated and the residue was dissolved in CH2CI2. After the solution was washed with 0.1 N HC1, the aqueous fraction was extracted with CH2CI2 (x 2), The organic fractions were combined, dried (Na2S04), and concentrated. The residue was purified by flash chromatography using CH2Cl2-20% MeOH in CH- L as eluents to obtain compound 86. 1H-NMR (400 MHz, CDC13) δ 12.02 (s, 1H), 10.40 (t, J = 5.7 Hz, 1H), 8.35 (s, lH), 6.63 (t, J = 8.1 Hz, 2H), 4.62 (d, J - 5.7 Hz, 2H), 4.59 (s, lH), 4.22 (dd, J = 12.2, 3.5 Hz, 1H), 4.13 (t, J = 1 1.9 Hz, 1H), 4.05 (dt, J - 12.0, 3.1 Hz, 1 H), 2.77 - 2.70 (m, 1 H), 2.31 m, 1H), 2.09 - 1.93 (m, III), 1.93 - 1 .81 (m, 2H), 1.10 (ddd, J = 13.9, 5.0, 2.1 Hz, 1 H), 1 .02 (d, J = 6.9 Hz, 3H). WF-NMR (376 MHz, CDC13) δ -109.22 (ddd, J 15. 1 . 8.7, 6.1 Hz, I F), - 1 12.05 (t, J = 6.9 Hz, 2F). LCMS-ESI+ ijn/z): [M +H]+ calculated for ( \-f S ·. ; I· ,' .-() ,: 448.2; found: 448.3.
Example 87
Preparation of ,/,v-5~ammotetrahydro-2H~pyran
Figure imgf000184_0001
Step 1
A solution of benzyl (5-oxotetrahydro-2H-pyran-3-yl)carbamate (740 mg, 3.0 mmol) and cerium(IH) chloride heptahydrate (1.12 g, 3.0 mmol) in 20 mL methanol was cooled to 0 °C and sodium borohydride (120 mg, 3.2 mmol) was then added poriionwise. The reaction mixture was allowed to stir at 0 °C for 45 minutes and then quenched by slow addition of 1 mL acetone followed by 3 hours stirring at room temperature. The reaction mixture was partitioned between water and dichloromethane and the aqueous phase extracted into dichloromethane followed by 2-butanol. The combined organic phases were dried over magnesium sulfate, filtered, concentrated, and the residue purified by flash chromatography (0-100% EtOAc/hexanes) to afford the desired cw-benz l ((3R,5S)-5-hydroxytetrahydro-2H~pyran-3-yl)carbamate. 1H- NMR (400 MHz, Chloroform-*/) 6 7.39 - 7,26 (m, 5H), 6,06 (br s, ! ! ! }. 5 07 (s, 2H), 3.86 - 3,70 (m, 2H), 3 ,69 - 3.47 (m, 4H), 2.00 - 1.89 (m, 1 H), 1 ,76 (d, ,/ 13.5 Hz, 1 H). The undesired tnms-isomer was also isolated. Step 2
To a solution of cz's-benzyl ((3R,5S)-5-hydroxytetrahydro-2H-pyraii-3- yl)carbamate (290 mg, 1.16 mmol) in 5 mL 1 : 1 DCM:EtOH was added 10wt% Pd/C (255 mg). This mixture was stirred under balloon pressure hydrogen for 18 hours and palladium removed by filtration thru celite with ethanol rinse. Upon concentration of filtrate, the c2s-5-ammotetrabydro-2H-pyran-3-ol was afforded and carried on as crude.
Example 88
Preparation of Compound 88
X2R,5SJ3aR)-N-(3-chioro-2-fiuoiObeiizy^
octahydro-2,5-methanopyrido[ 1 ',2':4,5]pyrazino[2, 1 -b][ 1 ,3]oxazepine- 10-carboxamide
Figure imgf000185_0001
88
Compound 88 was prepared in a similar manner to compound 15 using (3-chloro-2-fluorophenyl)methanamme in place of (4-fluoropheny!)methanamine. 3H- NMR (400 MHz, Chloroform-i/) δ 10.43 (br s, 1H), 8.34 (br s, 1H), 7.32 - 7.24 (m, 2H), 7.02 (t, J = 7.9 Hz, 1H), 5.36 (d, J = 9.4 Hz, 1H), 5.30 (s, 2H), 4.70 (d, J = 6.0 Hz, 3H), 4,24 (d, ./ 12.0 Hz, ! ! ! }. 4.00 (dd, J = 12.7, 9,5 Hz, 1H), 2.18 - 1.96 (m, 4H), 1 .96 - 1.83 (m, 1 H), 1 .60 (dt, J = 12.4, 3.1 Hz, i l l ). LCMS-ESI+ (m/z) \ XI I I I calculated for C21Hi9ClFN305: 448.1 1 ; found: 448.2. Example 89
Preparation of Compound 89
(2R.5S, 13aR)-N-(2,5-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,l 3, 13a- octahydro-2,5-methanopyrido[l\2':4,5]pyrazmo[2, l -b][l ,3]oxazepme-l 0-carboxamide
Figure imgf000186_0001
89
Compound 89 was prepared in a similar manner to compound 15 using (2,5-difluorophenyi)methanamine in place of (4-fluorophenyl)methanamine. !H- MR (400 MHz, Chloroform-,: ) δ 10.32 (t, 5.8 Hz, IH), 8.31 (br s, IH), 7.15 - 6.89 (m, 2H), 6.86 (cL ./ 8.5 Hz, I H), 5.40 (d, J = 9.3 Hz, I I I ). 5.24 (s, I H), 4.67 - 4.51 (m, 3H), 4.35 - 4.28 (m, I H), 3.99 - 3.90 (m, IH), 2.16 - 1.85 (m, 5H), 1.60 - 1.50 (m, IH). LCMS-ESI (m/z): [M+Hf calculated for C21H19F2N3O5: 432.14; found: 432.2.
Example 90
Preparation of Compound 90
(lR,4S42aR)-N-(3-chioro-2-fl orobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12a- octahydro-l ,4-methanodipyrido[l ,2-a: r,2'-d]pyrazme-9-carboxamide
Figure imgf000186_0002
Compound 9Θ was prepared in a similar manner to compound 41 using (3-ch.ioro-2-fluorophenyi)methanamme in place of (2,4,6-trifluorophenyl)methanamine. 1H-NMR (400 MHz, Chloroform-a') 6 9.22 (s, I H), 8.79 (s, IH), 7.39 - 7.28 (m, 2H), 7.06 (t, J = 8.0 Hz, I H), 4.89 (s, IH), 4.70 - 4.56 (m, M l ). 4.06 - 3.83 (m, 2H), 3.04 2.88 (m, 1H), 2.77 (s, III), 1.97 - 1.58 (m, 6H). LCMS-ESI+ {m/z): [M ill calculated forC2iHi9ClFN304: 432.11; found: 432.2.
Example 91
Preparation of Compound 91
(lR,4S,12aR)-7-hy(koxy-6,8-d^
octahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazme-9-carboxamide
Figure imgf000187_0001
Compound 91 was prepared in a similar manner to compound 41 using (2,3,4-trifluorophenyl)meth.anamine in place of (2,4,6-trifluorophenyl)met anamme. 1H-NMR (400 MHz, Chloroform-*) δ 10.25 (s, ill), 8.45 (s, ill), 7.10 id. J = 5.1 Hz, IH), 6.90 (d, ./' 8.7 Hz, IH), 4.89 (s, ill).4.63 (s, 2Π).4.22 (d, J= 11.6 Hz, IH), 3.93 - 3.73 (m, 2H), 2.71 (s, IH), 1.97 - 1.57 (m, 6H). LCMS-ESI+ (m/z): \ VI -Hi calculated for (\.H: J\»\';(>,: 434.13; found: 434.2.
Example 92
Preparation of Compound 92
(1 R,4S, 12aR)-N-(4-chloro-2-fluorobenzyi)-7-hydroxy-6,8-dioxo-l ,2,3,4,6,8,12,12 a- octah dro-l.,4-metb nodΐpyrido[l52-a:] 2'-d]pyrazme-9-carboxamide
Figure imgf000187_0002
Compound 92 was prepared in a similar manner to compound 41 using (4~chloro~2-fluorophenyl)methanamme in place of (2,4,6-trifluorophenyl)m.ethanam.ine. 'H- MM (400 MHz, Chloroform-*/) δ 10.28 (s, lH), 8.41 (s, III), 7.29 (s, 1H), 7.11 - 6.95 (m, 2H), 4.85 (s, 111).4.57 (s, 2H), 4.22 (d, J = 10.2 Hz, 1H), 3.81 (q, J = 13.9, 13.1 Hz, 2H), 2.68 (s, 1H), 1.99 - 1.50 (m, 6H). LCMS-ESI+ (m/z): |V5 lij calculated for C2iH19ClFN304: 432.11; found: 432.2.
Example 93
Preparation of Compound 93
(lR,4S,12aR)-N<2-chloro-4,6-dtf
octahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazme-9-carboxarnide
Step 1
A 5 mL microwave vial was charged with 2-bromo-l-chioro-3,5- difluorobenzene (540 mg, 2.4 mmol), cuprous cyanide (436 mg, 4,87 mmol), tetrakis(triphenylphosphme)palladium (63 mg, 0.05 mmol), sealed, and evacuated/backfilled with nitrogen. To this was added 5 mL degassed DMF. The sealed vessel was heated at 110 °C for 18 hours, diluted with ethyl acetate, and washed sequentially with twice 9:1 NH40H:NH4Ci aq twice 5% LiCl(aq ), and brine. The organic phase was then dried over magnesium sulfate, filtered, and concentrated. The crude residue was purified by flash chromatography (100% hexanes) to afford 2-chloro-4,6- difiuorobenzonitrile. ¾- MR (400 MHz, Chloroform-*/) δ 7.13 (dt, J ------- 8.0, 1.9 Hz,
1H), 6.93 (td, J = 8.5, 2.3 Hz, 1H).
Step 2
To a solution of 2-chloro-4,6-difluorobenzonitrile (210 mg, 1.2 mmol) in
2.4 mL THF was added a 2M solution of borane-DMS in THF (0.6 mL). This reaction mixture was allowed to stir at refiuxing temperature for 18 hours resulting in a loss of all solvent. The residue was re-dissolved in 3 mL THF, cooled to 0 °C, a 6M solution of HCl(aq) was carefully added, and the mixture returned to reflux for 30 minutes. The reaction mixture was once again cooled to 0 °C and treated with 4M NaOH(an . The aqueous phase was extracted with DCM, combined organic phases dried over magnesium sulfate, filtered, and concentrated. The crude residue was purified by flash chromatography (0-10% MeQH/DCM) to afford (2-chloro-4,6~ difluorophenyl)methanamine. ^l-N R (400 MHz, Chloroform-*/) 6 6.95 (dt, ,/ 8.3, 2.1 Hz, 1 1 1 ). 6.76 (td, ./ 9.4, 2,5 Hz, I I I }. 3.94 id. J 1 .9 Hz, 2H).
Steps 3 and 4
A solution of 93-A (74 mg, 0.1 1 mmol), (2-chloro-4,6- difluorophenyl)methanamine (48.5 mg, 0.27 mmol), HATU (100 mg, 0.26 mmol), and N.N-diisopropylethylamine (0.1 mL, 0.57 mmol) in 1 mL dichloromethaiie was stirred at room temperature for one hour at which point complete disappearance of 93-A and formation of 93-B was observed by LCMS. TFA (0.65 M) was added and the mixture was stirred at room temperature for one hour, at which point 1 mL DMF was added. The reaction mixture and then concentrated and purified by preparative HPLC (ACN/H20 + 0.1 % TFA) to afford compound 93. ¾- MR (400 MHz, DMSO-</6) δ 10.41 (t, ./ 5.7 Hz, 1 H ), 8.33 (s, 1 H), 7.41 - 7.26 (m, 2H), 4.72 - 4.57 ( rn. 3H), 4.43 (dd, J = 12.5, 3.6 Hz, 1H), 3.94 (t, J = 12.4 Hz, 2H), 3.77 (dd, J = 12.4, 3.6 Hz, 3H), 1.87 - 1.67 (m, 3H), 1.67 - 1.45 (ni, 2H), 1.43 (d, J = 10.4 Hz, 1H). LCMS-ESI* (ni/z): [M+H calculated for C r.. \ \ v..C\\- \ -£) 450.10; found: 450.2.
Preparation of Compound 94
(1 R,4S,12aR)-N-benzyl-7-hydroxy-6,8-dioxo-l ,2,3,4,6,8,12, 12a-octahydro-l ,4- methanodipyrido[ 1 ,2-a: 1 *,2'-d]pyrazine-9-carboxamide
Figure imgf000190_0001
Compound 94 was prepared in a similar manner to compound 41 using phenyl methanamine in place of (2,4,6-tri.fl.uorophenyl)methanamme. 1H- MR (400 MHz, Chloroform-*/) δ 10.37 (s, 1H), 8.26 (s, 1 H), 7.37 - 7.19 (m, 5H), 4.55 (d, J = 4.8 Hz, 1H), 4.34 (d, J = 5.7 Hz, 1H), 4.23 (d, J = 9.8 Hz, 1 H), 4.09 (d, J = 28.2 Hz, 1H), 3.78 (d, J = 10.9 Hz, I H), 3.64 (d, J = 13.2 Hz, 1 H), 3.14 - 3.01 (m, 1H), 1.91 - 1.49 (m, 4H). LCMS-ESI+ (ni/z) [M+H]+ calculated for C2,H21N304: 380.16; found: 380.2.
Example 95
Preparation of chiral tert-butyl 3-((l ,3-dioxoisoindoiin-2~yl)methyl)-2- azabicyclo[2.1.1 ]hexane-2-carboxylates 95- A and 95-B
Figure imgf000191_0001
Figure imgf000191_0002
Absolute stereochemistries unknown
Step 1
To a 0 °C solution of racemic tert-butyl 3 -(hydroxymethy l)-2- azabicyclo[2.1.1]hexane-2-carboxylate (285 mg, 1 .34 mmol), triphenylphospbine (425 mg, 1.62 mmol), and phthalimide (240 mg, 1.62 mmol) in 9 niL THF was added dropwise a solution of diisopropyl azodiearboxylate (0.35 mL, 1.8 mmol) in 1 ml THF. The reaction mixture was warmed to room temperature, stirred for 90 minutes, concentrated onto silica, and purified by flash chromatography (0-25% EtOAc/hexanes) to afford tert-butyl 3-((l 53-dioxoisoindolm-2-yl)meth l)-2-azabicyclo[2.1.1 ]hexane-2~ earboxylate as a racemic mixture. LCMS~ESI+ (m/z): [M+H] calculated for C i ,] ! -.\ .{ } ; : 343.2; found: 342.8.
Step 2
Racemic tert-butyl 3-((l ,3-dioxoisoindolm-2-yl)methyl)-2- azabicyclo[2.1.1 ]hexane-2-carboxylate (655 mg, 1.91 mmol) was separated by chiral FIPLC on a Lux Celluiose-2 column using an acetronitrile eluent to afford chiral 95-A (first eiuting peak) and 95~B (second eiuting peak) in enantioenriched form. For 95-A: 144 mg, 98%ee (absolute stereochemistr unknown). For 95-15: 242 mg, 49%ee (absolute stereochemistry unknown).
Example 96
Preparation of Compound 96
(1R,3R, 1 laS 6-hydroxy-5,7-dioxo-N-(2,4,6~trifluorobenzyl)-2,3,5
hexahydro-l H~l ,3-rnethanopyrido
Figure imgf000192_0001
Figure imgf000192_0002
Figure imgf000192_0003
Figure imgf000192_0004
Step 1
To a solution of intermediate 95-A (141 mg, 0.41 mmol, 98% ee, unknown absolute stereochemistry) in 9 mL ethanol was added hydrazine hydrate (0,5 niL, 10.3 mmol) and stirred at 70 °C for 18 hours to afford 96~A of unknown absolute stereochemistry. Solids were removed by filtration and the filtrate concentrated and carried on as crude. Step 2
A mixture of cmde 96-A (0.41 mmo! assumed), 96-B (430 mg, 1.25 mmol ), and sodium bicarbonate (69 mg, 0.82 mmol) in 2 mL water and 2 mL ethanol were stirred at room temperature for 18 hours, after which the reaction mixture was diluted with water and thrice extracted to ethyl acetate. The combined organic phases were dried over magnesium sulfate, filtered, concentrated. The crude residue (222 mg) was dissolved in 1.5 mL DCM and 4 N HCl in dioxane (4 mL) was added and stirred for 90 minutes at room temperature. The mixture was concentrated to dryness and coevaporated with toluene. The crude residue and DBU (0.3 mL, 2.0 mmoi) in 6 mL methanol was stirred at 50 °C for 90 minutes. The reaction mixture wras then concentrated onto silica gel and purified by flash chromatography (0-10% MeOH/DCM) to afford 96-C. LCMS-ESI+ i n/z): [M+H]+ calculated for C22H22N2O5: 395.16; found: 395.2.
Step 3
A mixture of 96-C (112 mg, 0.28 mmol), 1 M aqueous potassium hydroxide (1 mL), 4 mL methanol, and 4 mL THF was stirred at room temperature for 3 hours, at which point the mixture was diluted with dichloromethane, acidified by addition of 1M aqueous hydrogen chloride, and the organic phase extracted to dichloromethane. The combined organics were dried, filtered, and concentrated from toluene. After drying under vacuum, the residue was suspended in 1.5 mL DCM and trifluorobenzyiamine (62 mg, 0.38 mmol), HA.TU (220 mg, 0.58 mmol), and N,N~ diisopropyiethyl amine (DIPEA) (0.15 mL, 0.86 mmol) were added. This reaction mixture wras stirred at room temperature for 2 hours to afford 96-D which was carried forward as crude.
Step 4
Trifluoroacetic acid (1.7 mL, 22,2 mmol) was added to the crude reaction solution containing 96-D from the prior step and the reaction mixture allowed to stir at room temperature for 90 minutes. 1 mL of DMF was then added, the reaction mixture concentrated down to ~1 mL, filtered, and purified by preparative HPLC (ACN/water + 0.1% TFA) to afford compound 96 (unknown absolute stereochemistry). 'll-NVIR (400 MHz, DMSO-<¼) ό 10.45 - 10.35 (m, IH), 8.39 (s, IH), 7.23 - 7.09 (m, 2H), 4.67 (dd. J = 12.6, 4.8 Hz, 2H), 4.53 (d, J = 5.5 Hz, 2H), 4.20 (dd, J = 11.9, 3.8 Hz, IH), 4.05 - 3.95 (m, ill).2.96 - 2.88 (m, IH), 2.16 (d, J = 7.0 Hz, IH), 1.97 (d, J 7.0 Hz, IH), 1.68 - 1.60 (m, IH), 1.53 - 1.45 (m, IH). LCMS-ES ijn/z): \ VI II] calculated for ( ,.Ηι,Ι-'.-Ν^Ο,: 420.12; found: 420.2.
Example 97
Preparation of Compound 97
(1S,3S,11.aR)-6½di xy-5J-dioxo-N-(2,4,6-trifluorobenzyl)-2 ,5 J 1,11a- hexahydro-lH-l,3-methanopyrido[l ,2~a]pyrrolo[l,2~d]pyrazme~8~carboxamide
Figure imgf000194_0001
(Absolute stereochemistry unknown)
97
Compound 97 (49% ee, unknown absolute stereochemistry) was prepared in a similar manner to compound 96 using intermediate 95~B (49% ee, unknown absolute stereochemistry) in place of enantiom erica lly opposite intermediate 95-A. 'H-NMR (400 MHz, DM8C 6) δ 10.39 (t, J= 5.7 Hz, IH), 8.42 (s, IH), 7.25 - 7.13 (m, 2H), 4.73 - 4.66 (m, 2H), 4.54 (d, J = 5.7 Hz, 2H), 4.20 (dd, J= 12.3, 3.9 Hz, IH), 4.01 (t, J= 12.4 Hz, IH), 2.93 (dd, J= 6.7, 3.4 Hz, IH), 2.19 - 2.14 (m, IH), 1.97 (d, J= 8.3 Hz, IH), 1.65 (dd, J= 10.4, 7.9 Hz, IH), 1.49 (dd, J= 10.5, 7.7 Hz, IH). LCMS-ESr (m/z): [M+H]+ calculated for C2oHi6F3N304: 420.12; found: 420.2.
Example 98
Preparation of Compound 98
( 18,4Pv, 12aR)-3,3-difluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)- l,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[i,2-a:l 2'-d]pyrazine-9- carboxamide
Figure imgf000195_0001
Figure imgf000195_0002
Step 1
98-A (0.5g, 1 ,87mmol) was dissolved in DCM (20 mL) and cooled to 0 °C under Nitrogen, Dess-Martin Periodinane (1.59 g, 3.74 mmol) was added slowly. The mixture was stirred for 2 h at room temperature, quenched with Na2S203/NaHC03 (7: 1) aqueous saturated solution (160 mL) and stirred vigorously until two layers were separated. The cmde product was twice extracted with DCM. The combined organic iayers was dried over sodium sulfate and concentrated. The crude product was purified by flash chromatography on silica gel with 0-20%MeOH/DCM to afford 98-B. 3H- MHz, Chloroform-d) δ 4.34 - 4.05 (m, 1H), 3.97 - 3.75 (m, 1H), 3.69 (s, M i l 2.89 (dd, J = 4.4, 2.1 Hz, ! ! ! }. 2.30 - 1.97 (m, 3H), 1.56 (d, J = 11.3 Hz, ! ! ! }. 1.35 (s, 9H). LCMS-ESI+ (m/z) [M+H]'4 calculated for C13Hi9N05: 269.13; found: 270.78.
Step 2
A solution of 98-B (504 mg, 1.87mmo!) in DCM (15 mL) was stirred at
0 °C. DAST (1ml) was added drop wise to the reaction mixture. After overnight stirring at room temperature, the reaction mixture was cooled back to 0 "C. Saturated NaHC0 (10 mL) was added slowly. The mixture was extracted with twice with DCM and dried over Na2S0 . After concentrating, the residue was purified by flash chromatography 0-50% EtOAe/hexanes to give 98-C. 1H-NMR (400 MHz, Chloroform-d) δ 4.45 - 4.18 (m, 1 H), 3.85 (m, i l l ). 3.72 (d, .1 - 1.5 Hz, 3H), 2.72 (ddd, J = 5.1, 3.2, 1.6 Hz, 1 1 1 2.27 - 1.52 (m, 4H), 1.41 (d, J = 21.9 Hz, 9H). 19F-NMR (376 MHz, Chloroform-d) δ -91.72 - -93.99 (m), -113.65 - -115.98 (m). LCMS-ESF ijn/z): [M+H calculated for CDH^ 291.13; found: 291.55.
Step 3
98~C (476 mg, 1 .634mmol) in THF (20 mL) was stirred at 0 °C as 2.0 M LiBl:L in THF (2.4 mL, 4.8mmof) was added. The mixture was warmed to room temperature and stirred for 4 h. The reaction mixture was quenched with ice and diluted with EtOAc and saturated NH4CI (some H2 evolution). After the two phases were separated, the organic fraction was washed with brine, dried a2S04), and concentrated. The crude product of 98-D was used as is for the next step, LCMS-ESI+ (m/z): [M+H]'1' calculated for C12H19F2NQ : 263.13; found: 164.10. Step 4
98-D ( 1.634mmoi), phthaiimi.de (0.36 g, 2.4 5mmol ), and PPh3 (0.855 g, 3.26mmol) in THF (10 mL) was stirred at 0 °C bath as DIAL) (0.642 mL, 3.26mmol) was added. After addition, the mixture was stirred at 0 °C for 30 min and then at room temperature for 16 h. It was diluted with EtOAc, and saturated NH4CI. After stirring for 5 min, a solid was filtered off and the two phases were separated. The organic phase was washed with brine, dried (Na2SQ4), and concentrated. The crude product was purified by flash, chromatography with 0-50%EA/Hex as eiuents to give 98-E. 1H- NMR suggests a mixture of two rotarners. H-NMR (400 MHz, Chloroform-d) δ 7.89 - 7.80 (m, 2H), 7.78 - 7.66 (m, 2H), 5.02 (ddt, J - 16.6, 12.5, 6.3 Hz, IH), 4.24 (d, J - 71.8 Hz, IH), 4.10 - 3.92 (m, 1 H), 3.83 - 3.51 (m, 2H), 2.46 (s, 1H), 2.21 - 1 .98 (m, 2H), 1 .87 - 1.62 (m, 2H), 1 .31 (d, J - 8.5 Hz, 9H); WF-NMR (376 MHz, Chloroform-d) δ -91 .22 - -93.58 (m), -1 13.20 - -1 15.45 (m). LCMS-ESI+ (m/z): | VI - H i calculated for ( · ,,Η . .!: .-\· .() . : 392.15; found: 393.3.
Step 5
To a solution of 98-E (696 mg, 1.774mmol) in EtOH (lOmL) was added hydrazine hydrate (ImL) at room temperature and the resulting solution was stirred at room temperature for 2 h. The mixture was diluted with ethyl ether (30 mL) and stirred at 0 °C for 60 min before filtration. The filtrate was concentrated and the residue was dissolved in CH2C12 and filtered. The filtrate was concentrated and purified by flash chromatography on silica gel with 0-20% MeOH (0.2% TEA) /DCM to give 9S~F, 1H~ NMR (400 MHz, Chloroform-d) 8 4.91 (p, J = 6.2 Hz, IH), 4.29 - 3.97 (m, IH), 3.36 - 2.93 (m, 2H), 2.49 (qt, J - 8.8, 5.2 Hz, 2H), 2.08 (dddd, J = 25.5, 14.0, 7.1 , 4.9 Hz, IH), 1 .89 - 1 .49 (m, 4H), 1.41 and 1.21 (d, J = 6.2 Hz, 9H). 19F-NMR (376 MHz, Chloroform-d) δ -91 .63 - -93.16 (m), -1 13.1 1 - -1 15.08 (m). LCMS-ESI* (m/z): [M+Hf calculated for ( -i j ,.] s■(> ·. 262.15; found: 262.8.
Step 6, 7 and 8
The mixture of 98-G (375.8 mg, 1.55 mmol), 98-E (370 mg, 1.41 mmol), and NaHCO^ (261 mg, 3.10 mmol) in water (5 ml.) and EtOH (5 mL) was stirred at room temperature for 2 h. The mixture was diluted with brine and extracted with EtOAc (x 2). The extracts were combined, dried (Na2S04), concentrated, and dried in vacuo to afford crude A. LCMS-ES (m/z): [M+H]+ 591 .59. Crude A (T .38mmol) in Cf S .{ 'I■ (5 mL) was added 4 N HC1 in dioxane (5 mL). After 2 h at room temperature, mixture was concentrated to dryness. It was co-evaporated with toluene once and dried in vacuo to afford crude B. B (1.38mmol + 0.442mmol) and DBU (3 mL, 1 lmmol) in anhydrous MeOH (15 mL) were stirred at 50 °C bath for 40 min. The mixture was concentrated. The residue was purified by flash chromatography (80 g column) using 0 - 20% MeOH/DCM as eluents to give 98-H. LCMS-ESI+ (mz): [M+HJ+ calculated for C23H22F2 2O5: 44415: found: 445.36 (90%), 431.18 (10%).
Steps 9,10 and 11
The remaining steps were performed using procedures similar to
Example 41 to afford desired compound 98. 1H-NMR (400 MHz, Chloroform-d) δ 10.29 (d, J = 6.1 Hz, 1H), 8.34 (s, 1H), 6.65 (dd, J = 8.7, 7.5 Hz, 2H), 4.83 (s, 1H), 4.72 - 4.58 (m, 2H), 4.36 - 4.10 (m, 2H), 4.05 ft, J - 11.5 Hz, 1H), 2.97 (d, J = 4.4 Hz, 1H), 2.49 - 2.08 (m, Ml).2.12 - 1.94 (m, 1H).19F-NMR (376 MHz, Chloroform-d) δ -92.08 - -93.57 (m, IF), -108.92 (ddd, J = 15.1, 8.8, 6.3 Hz, IF), -109.30 - -110.65 (m, IF), - 112.16 (p, J = 7.3 Hz, 2F). LC S-ESI+ (m/z): \ II] calculated for (·..:!!,<.!··-- 469.11; found: 470.23.
Example 99
Preparation of Compound 99
( 1 R.3S,4R.12»R )-7-hyciroxy-3-mci yl-6.8-dioNo 2. . - nuor<)bc! l )-
1 ,2,3,4,6,8, 12,12a-octahydro- 1 ,4-rnethanodipyrido[ 1 ,2-a: 1 ',2'-d]pyrazine-9- car box amide
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000199_0002
Step 1
To a stirred solution of 99-A (1 g, 3.71 mmoi) in THF (20 mL) was added dropwise a solution of the Tebbe reagent (0.5 M in toluene, 14.85mL, 7.42 mmol) at 0 °C. After addition, the brown solution was allowed to warm to room temperature slowly and was stirred at room temperature for 2 h. The reaction was quenched carefully by the addition of saturated NaHC€>3 solution at 0 °C, and the mixture was stirred at room temperature for 15 minutes. The mixture was filtered through ceiite, and the filter cake was washed with ether and DCM (1 : 1) twice. After separated layers, the organics were combined and concentrated in vacuo, and the residue was purified by column chromatography on silica gel column with 0-50% EtOAc hexanes to afford 99-B. !H-NMR (400 MHz, Chloroform-d) δ 5.06 (dt, J = 48.6, 2.6 Hz, 1H), 4.73 (d, J = 7.0 Hz, 1H), 4.42 (d, J = 61.8 Hz, 1H), 3.81 (d, J = 48.2 Hz, 1H), 3.73 (d, J = 1.6 Hz, 3H), 2.74 (dd, J = 9.4, 4.4 Hz, 1H), 2.38 (ddt, J = 13.5, 4.5, 2.5 Hz, 1 H), 2.18 - 2.06 (m, 1 H), 1.99 (dt, J = 10.2, 2.4 Hz, 1H), 1 .58 (s, 1H), 1.42 (d, J = 25.5 Hz, 9H). LCMS-ESI* (m/z): [M+H] ; calculated for CHH2IN04: 267.15; found: 267.65.
Step 2
A mixture of 99-B (675 mg, 2.506 mmol) and 20% Pd(OH)2/C (500 mg) in EtOH (50 mL) was stirred under H2 atmosphere. The mixture was filtered through Celite and the filtrate was concentrated to give 99-C. 1H-NM (400 MHz, Chloroform- d) δ 4.23 - 3.99 (m, H I ). 3.77 - 3.64 (m, 4H), 2.55 (d, J = 4.8 Hz, I H), 2.14 - 1 .86 (m, 3H), 1.42 (d, J = 24.2 Hz, 9H), 0.96 (d, J = 6.6 Hz, 3H), 0.85 (ddd, J = 12.5, 4.8, 2.4 Hz, 1 H). LCMS-ESJT (m/z): \ \\ i i j calculated for C , ;] ί, Ο ; . 269.16; found: 269.69.
Step 3
99-C (670 mg, 2.488 mmol) in THF (20 mL) was stirred at 0 °C as 2.0
M LiBH4 in THF (3.7mL, 7.46 mmol) was added. The mixture was warmed to room temperature and stirred for 4h, The reaction mixture was quenched with ice and diluted with EtOAc and saturated NH4CI (some H2 evolution). After two phases were separated, the organic fraction was washed with brine, dried (Na2S04), and concentrated. The crude alcohol 99-D was used as is for the next step. LCMS-ES (m/z): [M+Hf calculated for C0H23 O3: 241.17; found: 241 .76.
Steps 4 and 5
Steps 4 and 5 were performed using procedures similar to those in Example 41 to afford 99~F. LCMS-ES1+ (m/z): [M+H] calculated for ( " ; j !.■ ,: 240.18; found: 241.2.
Step 6, 7 and 8
Steps 6, 7 and 8 were performed using procedures similar to that of Example 41 to give 99-G. LCMS-ESI+ (m/z): [M+H]+ calculated for C24H26N205: 422.18; found: 423.21.
Step 9, 10 and 1 1 The remaining slops were performed using procedures similar to Example 41 to afford compound 99. ^I-NMR (400 MHz, Chloroform-d) 6 1 1.71 (s, 1 H), 10.36 (t, J = 5.7 Hz, IH), 8.28 (s, H i ). 6.63 (t, J = 8.1 Hz, 2H), 4.63 (t, J - 5.4 Hz, 3H), 4.12 (dd. J - 12.3, 3.5 Hz, IH), 3.83 (t, J - 12.3 Hz, IH), 3.67 (dd, J = 12.3, 3.4 Hz, I H), 2.64 - 2.52 (m, IH), 2.30 (ddq, J = 10.5, 7.2, 3.6 Hz, I H), 2.13 (td, J = 12.1 , 4.4 Hz, IH), 1.82 - 1.63 (m, 2H), 1.24 (d, J = 3.3 Hz, IH), 1.04 (d, J = 6.9 Hz, 4H), 0.90 - 0.79 (m, IH). 19F~ MR (376 MHz, Chloroform-d) 8 -109.20 (ddd, J = 15.0, 8.8, 6.2 Hz), -1 12.03 (t, J = 7.0 Hz). LCMS-ESI+ (m/z): [M+H] ! calculated for C22H2oF3N304: 447.14; found : 44832.
Example 100
Preparation of Compound
( 1 R,4R, 12aS)-N-(2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo- 1 ,2,3 ,4,6,8, 12, 12a- octahydro- 1 ,4-ethanodipyrido[ 1 ,2-ai 1 ',2'-d]pyrazine-9-carboxamide
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000202_0002
100-F 100-G
Figure imgf000202_0003
100
Step 1
A 100-mL rbf was charged with 100-A (2.0 g, 7.8 mmol) in THF (20 mL). The reaction mixture was cooled to 0 °C. Borane dimethyl sulfide (2 N in THF, 17.6 mL) was slowly added in. Then the reaction mixture was stirred at room temperature for overnight. The reaction mixture was cooled back to 0 °C. Methanol (8 mL) was added drop wise to quench the reaction. After concentration, the residue was purified by Combi Flash (40 g column, cartridge used) using hexanes - EA as eiuents to afford 100-B. I .C.VIS-KSI (m/z): j \1 · l l | found: 242.
Step 2
A 100-mL rbf was charged with 100-B (1.8 g, 7.4 mmol), triphenylphosp ine (4,3 g, 16.2 mmol) and phthalimi.de (1.8 g, 12.2 mmol) in THF (30 mL). Then the reaction mixture was cooled to 0 °C with stirring. D1AD (3.2 mL, 16.2 mmol ) was slowly added to the reaction mixture. The reaction mixture was stirred at room temperature for overnight. After concentration, the residue was purified by Combi Flash (80 g column, cartridge used) using hexanes - EA as eluents to afford 100-C. LCMS-ESI* (m/z): | \<! H j found: 371.
Step 3
To a solution of 100-C (2.5 g, 6.8 mmol) in EtOH (50 mL) was added hydrazine monohydrate (1.7 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford 100-D. LCMS-ESI+ (m/z): [M+H found: 241 .
Step 4
A 100-mL rbf was charged with ΙΟΘ-D (1.6 g, 6.7 mmol) and 100-E (2.3 g, 6.7 mmol) in etbanol (30 mL). Sodium bicarbonate (1.2 g, 1 ,4 mmol) in water (30 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for overnight. The mixture was diluted with EA (200 mL) and washed with water (2 x). The aqueous fractions were extracted with EA (1 x), and the organic fractions were combined, dried (Na2S04), and concentrated. The crude Ιθθ-F was used for next step without further purification. LCMS-ESI (m/z): [M+H]T found: 569.
Step 5
A 100-mL rbf was charged with 100-F (3.7 g, 6.5 mmol) in 4 N HQ /dioxane (38 mL). Then the reaction mixture was stirred at room temperature for 1 hour. After concentration, 3.2 g intermediate was obtained. The intermediate and DBU (5.1 g, 33.8 mmol) were dissolved in toluene ( 100 mL). The reaction mixture was heated to 1 10l'C with stirring for 1 hour. After concentration, the residue was purified by Combi Flash (80 g column, cartridge used) using hexanes - EA as eluents to afford ΙΘΟ-G. LCMS-ESI"' (m/z): [M+H]+ found: 423. Step 6
A 100-mL rbf was charged with 100-G (2.0 g, 4.7 mmol) in THF (20 mL) and MeOH (20 mL). 1 N KOH (18.9 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for I hour. The reaction mixture wras acidified by adding 1 N HQ (18.9 mL). After concentration, the residue was co- evaporated with toluene (3 x). The crude acid (0.28 g, 0.72 mmol), 2, 4- difluobenzylamine (0.2 g, 1.44 mmol), Ν,Ν-diisopropylethylamme (DIPEA) (0.47 g, 3.6 mmol) and HATU (0.55 g, 1 .44 mmol) were dissolved in DCM (20 mL). The reaction mixture was stirred at room temperature for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated NaHC03 (2x), saturated NH4C1 (2x) and dried over Na2S04. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAe to afford 1ΘΘ-Η. LCMS-ESF (m/z): [M+H]+ found: 520. Step 7
A 50-mL rbf was charged with 100-H (0.36 g, 0.69 mmol) in TFA (5 mL). The reaction mixture was stirred at room temperature for 30 minutes. After concentration, the crude was purified by column chromatography on si lica gel with EtOAc-MeOH to afford compound 10Θ. ' H-NMR (400 MHz, Chloroform-d) δ 12.25 (m, 1 H), 10.47 (t, J = 5.9 Hz, 1H), 8.30 (s, I H), 7.58 - 7.29 (m, 1H), 6.98 - 6.50 (m, 2H), 4.62 (dd, J = 14.8, 4.9 Hz, 3H), 4.22 (t, J = 12.2 Hz, 1H), 4.14 - 4.07 (m, i l l 3.96 (dd, J = 12.2, 3.1 Hz, 1H), 2.26 - 1.44 (m, 9H). 19F-NMR (376 MHz, Chloroform-d) 8 - 1 12.38 (t, J - 7.7 Hz), -1 14.78 (q, J = 8.5 Hz). LCMS-ES (m/z): found: 430.
Example 101
Preparation of Compound 101
(1 R,4R, 12aS)-7-hydroxy-6,8-dioxo-N-(2,4,6-trifliiorobeTizyl)- 1 ,2,3,4,6,8, 12, 12a- octahydro- 1 ,4-ethanodipyrido[ 1 ,2-a: 1 ',2'-d]pyrazine-9-carboxamide
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000205_0002
A 100-mL rbf was charged with 101-A (0.3 g, 0.72 mmol) in THF (2 mL) and MeOH (2 mL). 1 N KOI I (2.1 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was acidified by adding 1 N HC1 (2, 1 mL). After concentration, the residue was co- evaporated with toluene (3 x). The cmde acid (0.72 mmol), 2, 4, 6-trifluobenzylamine (0.23 g, 1.44 mmol), N,N-diisopropyiethylamme (D1PEA) (0.47 g, 3.6 mmol) and HATU (0.55 g, 1.44 mmol) were dissolved in DCM (20 mL). The reaction mixture was stirred at room temperature for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated 'NaHC03 (2x), saturated M I (2x) and dried over Na2S04. After concentration, the cmde was purified by column chromatography on silica gel with hexane-EtOAc to afford 101-B. LCMS-ESI+ (m/z): [M+H found: 538.
A 50-mL rbf was charged with 101-B (0.36 g, 0.67 mmol) in TFA (5 mL). The reaction mixture was stirred at room temperature for 30 minutes. After concentration, the crude was purified by column chromatography on si lica gel with EtOAc-MeOH to afford compound 101. 1H-NMR (400 MHz, Chioroform-d) δ 12.1 1 (s, 1 H), 10.40 (t, J = 5.8 Hz, 1H), 8.28 (s, 1H), 6.91 - 6.39 (m, 2H), 4.62 (ddd, J = 25.0, 6.5, 2.8 Hz, 1 i h 4.21 (t, J - 12.2 Hz, !!!}.4.09 (dd, J - 12.5, 3.0 Hz, ill).3.93 (dd, J 12.2, 3.1 Hz, 1H), 2.35 - 1.39 (m, 9H).19F NMR (376 MHz, Chloroform-d) δ -112.3 (t, J = 7.7 Hz), -114.78 (q, .1 = 8.5 Hz). LCMS-ESI+ (ni/z): found: 448.
Example 102
Preparation of Compound 102
(lS,4S42aR)-7-hydroxy-6 -dioxo-N 2,4,6-trifluorobenzyl)-l,23,4,6^ 2J2a- octahydro- 1 ,4-et anodipyrido[ 1 ,2-a: 1 ',2'~d]pyrazine~9-carboxamide
Figure imgf000206_0001
Figure imgf000207_0001
32-E
Figure imgf000207_0002
Step 1
A 100-mL rbf was charged with 102- A (2.0 g, 7,8 mmol) in THF (20 mL). The reaction mixture was cooled to 0 °C. Borane dimethyl sulfide (2 N in THF, 17.6 mL) was slowly added in. Then the reaction mixture was stirred at room temperature for overnight. The reaction mixture was cooled hack to 0 °C. Methanol (8 mL) was added drop wise to quench the reaction. After concentration, the residue was purified by Combi Flash (40 g column, cartridge used) using hexanes - EA as eluents to afford 102-B. LCMS~E8I+ (ni/z): j .Vt H | found: 242. Step 2
A 100-mL rbf was charged with 102-B (1 .8 g, 7,4 mmol), triphenylphosphine (4,3 g, 16.2 mmol) and phthaiimide (1.8 g, 12.2 mmol) in THF (30 fflL). Then the reaction mixture was cooled to 0 °C with stirring. DIAD (3.2 mL, 16.2 mmol) was slowly added to the reaction mixture. The reaction mixture was stirred at room temperature for overnight. After concentration, the residue was purified by Combi Flash (80 g column, cartridge used) using hexanes - EA as eluents to afford 102-C. LCMS-ES (m/z) [M+H]+ found: 371.
Step 3
To a solution of 102-C (2.5 g, 6.8 mmol) in EtOH (50 mL) was added hydrazine monohydrate (1.7 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford 102-D. LCMS-ESI+ (m/z): [M+H]+ found: 241.
Step 4
A 100-mL rbf was charged with 102-D (1.6 g, 6,7 mmof) and 102-E (2.3 g, 6.7 mmol) in ethanol (30 mL). Sodium bicarbonate (1.2 g, 1.4 mmol) in water (30 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for overnight. The mixture was diluted with EA (200 mL) and washed with water (2 x). The aqueous fractions were extracted with EA (1 x), and the organic fractions were combined, dried (Na2S04), and concentrated. The crude 102-F was used for next step without further purification. LCMS-ESI4 (m/z): [ +H]+ found: 569.
Step 5
A 100-mL rbf was charged with 102-F (3.7 g, 6.5 mmol) in 4 N HC1 /dioxane (38 mL). Then the reaction mixture was stirred at room temperature for 1 hour. After concentration, 3.2 g intermediate was obtained. The intermediate and DBU (5.1 g, 33.8 mmol) were dissolved in toluene (100 mL). The reaction mixture was heated to 1 0°C with stining for 1 hour. After concentration, the residue was purified by Combi Flash (80 g column, cartridge used) using hexanes - EA as eluents to afford 102-G. LC S~ES (m/z): | VI · ! ! | found: 423.
A 100-mL rbf was charged with 102-G (0.3 g, 0.72 mmol) in THF (2 mL) and MeOH (2 mL). 1 N OH (2.1 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was acidified by adding 1 N HC1 (2, 1 mL). After concentration, the residue was co- evaporated with toluene (3x). The crude acid (0.72 mmol), 2, 4, 6-trifiuobenzylamine (0.23 g, 1.44 mmol), N,N-diisopropyiethylamme (DIPEA) (0.47 g, 3.6 mmol) and HATU (0.55 g, 1.44 mmol) were dissolved in DC (20 mL). The reaction mixture was stirred at room temperature for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated NaHC03 (2x), saturated NH4CI (2x) and dried over Na2S04. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford 102-H. LCMS-ESI* (m/z): [M+H]+ found: 538.
Step 7
A 50-mL rbf was charged with 102-H (0.36 g, 0.67 mmol) in TEA (5 mL). The reaction mixture was stirred at room temperature for 30 minutes. After concentration, the crude was purified by column chromatography on si lica gel with EtOAc-MeOH to afford compound 102. 1H-NMR (400 MHz, Chioroform-d) δ 12.13 (s, I H), 10.40 (t, J = 5.8 Hz, 1 H), 8.28 (s, i l l ). 6.64 (t, J = 8.1 Hz, 2H), 4.89 - 4.41 (m, 3H), 4.22 (t, J = 12.2 Hz, IH), 4.09 (dd, J = 12.3, 3.1 Hz, IH), 3.95 (dd, J = 12.1 , 4.1 Hz, IH), 2.45 - 1.60 (m, 9H). 19F-NMR (376 MHz, Chloroform-d) δ - 109.26 (ddd, J = 15.1 , 8.8, 6.3 Hz), -1 1 1.99 (t, J - 6.9 Hz). LCMS-ES m/z): found: 448.
Example 103
Preparation of Compound 103
(l R,4R, 12aR)-2,3-dit1uoro-7-hydroxy-6,8-dioxo~N-(2,4,6-tafiuoroben
1 , 2,3,4,6,8, 12 J.2a-octahydro- 1 , 4-methanodipyri.do[ 1 ,2-a: ,2'-d]pyrazine-9- carboxamide
Figure imgf000210_0001
Figure imgf000210_0002
103-A 103-B
Figure imgf000210_0003
Step 1
A solution of (l R,3R,4R,5R,68)-methyi 5,6~dihydroxy-2~((S)-l ~ phenylethyl)-2-azabicyclo[2.2.1]heptane-3-carboxylate (2.0g, 6.9mmol) in DCM (27mL) was cooled to -78 °C in a dry ice/acetone bath. To this solution was added DAST (2.18 mi, 16.48 mmol) via plastic tipped pipette. The solution was stirred at -78 °C for 30 minutes after which time it was removed from the bath, let warm slowly to room temperature, and stirred at room temperature for one hour. The reaction was quenched by slow addition of the reaction mixture to a stirring solution of saturated sodium bicarbonate (150mL) via plastic tipped pipette. The layers were separated and the aqueous layer was back-extracted with dichloromethane. The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (7-28% ethyl acetate/hexane) to provide 103-A. Ή-NMR (400 MHz, Chloroform-,: ) δ 7.43 - 7.16 (m, 5H), 5.01 - 4.60 (m, 2H), 3.85 (q, J ------- 7.1 , 6.6 Hz, 1H), 3.55 (s, 2H), 3.53 - 3.42 (m, 2H), 2.76 (dq,
J = 5.1 , 2.0 Hz, 1 H), 2.19 - 2.07 (m, 1H), 2.03 - 1.88 (m, 1 H), 1.39 (d, J = 6.7 Hz, 3H).
Steps 2. and 3
To a solution of 103-A (0.96 g, 3.24 mmol) in Ethanol (36.01 ml) and 1 .25M HCl-ethanol (4.09 ml) was added 20% PdOH/C (1.14 g, 1.62 mmol) the suspension was stirred under an atmosphere of hydrogen for 22 hours. After filtering through Celite, the cake was washed with EtOH, the filtrate was concentrated under vacuum to dryness to afford the crude deprotected product which was assumed to be 3.24mmol for next step. LCMS-ESI+ (m/z): [M+H]+ calculated for CJ ! ; >!·' ·Ν() 192.08; found: 192.1 10.
To the crude residue (0.62 g, 3.24 mmol) and Di-tert-butyl dicarbonate (1.06 g, 4.86 mmol ) in 2-Methyltetrahydrofuran (32.43 ml) was added N,N- diisopropylethyiamine (0.56 mi, 0 mol). Upon completion, the reaction mixture was diluted with water, extracted into EtOAC (2x) and the organic fractions were washed with water, combined, dried (Na2S04), and concentrated. The residue was purified by silica column chromatography (0-55% EtOAc/Hexanes) to afford 1Q3-B. !H-NM (400 MHz, Chloroform-d) δ 5.12 - 5.01 ( rn. Ill), 4.92 (s, I H), 4.49 (s, IH), 4.14 (d, J 14.7 Hz, I H), 3.75 (s, 3H), 2.91 (s, 1 1 1 ), 2.24 - 1.98 (m, 2H), 1 .47 (s, 51 1 ). 1.38 (s, 51 1 ). LCMS-ESf (m/z) [M+H]+ calculated for C13H20F2NO4: 292.13; found: 291.75.
Step 4
A solution of 103-B (0.68 g, 2,33 mmol) in THF (15 ml) was stirred in an ice bath as 1 .0 M Li.BH4 in THF (4.65 ml) was added and the resulting mixture was uStirred at 0 °C for 30 minutes at which time it was shown to be complete by TLC. The reaction mixture was carefully treated with water (0.3 mL), then with NaOH (-15%, 3.5M, 0.3 mL), then finally with additional water (0.9 mL). The mixture was stirred at room temperature for 15 minutes, and the ppt that formed was filtered, washed with diethyl ether and the supemate was concentrated to afford 103-C. 1H-NM (400 MHz, Chioroform-d) δ 4,83 (s, 1H), 4.56 (d, J = 10.5 Hz, 1H), 4.37 (s, 1H), 3.78 - 3.47 (m, M i l 2.76 (s, I H), 2.36 - 2.18 (rn. 1 H), 2.17 - 1.98 (m, I H), 1 .55 (s, I H), 1 .48 (s, 9H).
Steps 5 and 6
A mixture of 103-C (0.59 g, 2.25 mmol), phthalimide (0,53 g, 3.6 mmol) and triphenylphosphine (1.3 g, 4.95 mmol) in THF (1 1 ml) was cooled in an ice bath. Diisopropyl Azodicarboxyiate (0.97 ml, 4.95 mmof) was added. The mixture was then warmed up to room temperature and stirred for 14h and then concentrated in vacuo. The residue was dissolved in ether, stirred for 1 h, then the solids were filtered off and the filtrate was concentrated. The residue was purified by silica column chromatography (10-31-91% EtOAc/hexanes) to afford the protected amino compound (assumed 2.25mmoi of product). LCMS-ESI+ (m/z): [M+H]+ calculated for C20H23F2N2O4: 393, 15; found: 392.77.
A solution of the protected amino compound (0.88 g, 2.25 mmol) and hydrazine hydrate (0.46 ml, 9.52 mmol) in ethanoi (22 ml) was stirred at 60 °C for 2 h. The reaction mixture was cooled in an ice bath, ether (10 ml) was added and the mixture was stirred for 30 min. The solid formed was filtered off and the filtrate was concentrated under vacuum to dryness to give 103-D. 1H-N R (400 MHz, Chloroformed) 6 5.17 - 4.61 (m, 2H), 4.37 (s, IH), 3.80 (s, IH), 3, 1 1 - 2.77 (m, IH), 2.01 (s, 2H), 1 .87 (s, IH), 1.83 i d. J = 7.4 Hz, I H), 1 .46 (s, 9H), 1.30 (d, J = 6.4 Hz, IH), 1 ,27 (d, J = 6.3 Hz, 3H). LCMS-ESI+ (m/z): [ \1 I I ! calculated for C12H20F2N2O2: 263.15; found: 262.86.
Steps 7, 8 and 9
Compound 103 was prepared in a similar manner to compound 60 using 103-D in place of 41-E and using (2,4,6-trifluorophenyl)rneihanamine in place of (2,3- dichlorophenyl)methanamine. A single diastereomer resulted. The stereochemistry of the fluorines is unknown. 1H-NMR (400 MHz, Chloroform-d) δ 8.08 (s, I H), 6.46 - 6.27 (m, 2H), 4.95 (d, J = 53.5 Hz, IH), 4.65 (d, J = 54,9 Hz, IH), 4.45 (s, IH), 4,33 (d, J = 5.6 Hz, 2H), 3.84 (t, J = 3.6 Hz, 2H), 2.75 (s, 1 H), 2.28 (p, J = 1.9 Hz, 2H), 2.20 (s, 1H), 1.91 (dd, J = 33.3, 15.2 Hz, 1 H), 0.95 (s, 1 H). LCMS-ES (m/z): [M+H]+ calculated for C . : H H:« <> i: 470.1 1 ; found: .470.13. ANTIVIRAL ASSAY
Example 104
Antiviral Assays in MT4 Cells
For the antiviral assay utilizing MT4 cells, 0.4 Ε of 189 test concentration of 3-fold serially diluted compound in DMSO was added to 40 ,uL of cell growth medium (RP 1 1640, 1 0% FBS, 1 % pemciiline/Streptomycine, 1 % L- Glutamine, 1 % HEPES) in each well of 384-wel I assay plates (10 concentrations) in quidruplicate.
1 mL aliquots of 2 ¾ 106 MT4 cells are pre-infected for 1 and 3 hours respectively at 37 °C with 25 (uL (MT4) or of either cell growth medium (mock- infected) or a fresh 1 :250 dilution of an HlV-lIIb concentrated ABI stock (0.004 m.o.i. for MT4 cells). Infected and uninfected cells are diluted in cell growth medium and 35 μ,Ε of 2000 (for MT4) cells is added to each wel l of the assay plates.
Assay plates were then incubated in a 37 °C incubator. After 5 days of incubation, 25 Ε of 2X concentrated CellTiter-Glo™ Reagent (catalog # G7573, Promega Biosciences, Inc., Madison, Wl) was added to each well of the assay plate. Cell lysis was carried out by incubating at room temperature for 2-3 minutes, and then chemiluminescence was read using the Envision reader (PerkinElmer).
Compounds of the present invention demonstrate antiviral activity in this assay as depicted in Table 1 below. Accordingly, the compounds of the invention may be useful for treating the proliferation of the HIV virus, treating A I DS, or delaying the onset of A I DS or ARC symptoms.
Table 1
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Example 105
H masi PXR Activation Assay
Luciferase Reporter Gene Assay. A stably transformed tumor cell line ( DPX2) was plated on 96-well microliter plates. DPX2 cells harbor the human PXR gene (NR1I2) and a luciferase reporter gene linked to two promoters identified in the human CYP3A4 gene, namely XREM and PXRE. The cells were treated with six concentrations of each compound (0.15 ~ 50 μ,Μ) and incubated for 24 hours. The number of viable cells was determined and the reporter gene activity was assessed. Positive control : Rifampicin at 6 concentrations (0.1 ~ 20 μΜ). ( ]„..< relative to the maximum fold induction by 10 or 20 μΜ RIF was calculated for test compounds according to the following equation which adjusts for the DMSO background: %Emax = (Fold induction - l)/(Maximum fold induction by RIF - 1) x 100%.
Table 2
Figure imgf000218_0001
Compound %Emax at
Number 15 μΜ
40 0
41 11.5
42 21
4J 18
44 4
45 19
46 34
47 1 1
48 5
54 ->
55 24
56 3
57 3
58 1
59 4
60
61 1
63 13
64 8
66 0
67 0
68 6
69 5
70 10
71 3
72 4
73 7
75 0 Compound %Emax 3·ί
Number 15 μΜ
7 11
79 0
80
81 I
82 1
83 I
84 "> 1
85 77
86 30
88 27
89 5
90 11
91 3
92 3
93 9
96 11
97 9
98 0
99 17
100 45
102 123
103 0
Example 106
OCT2 Inhibition Assay
The dose dependent inhibition of OCT2 mediated uptake of a model substrate ¾4C-Tetraethylammoiiium (TEA) by test compounds was studied in wild-type and OCT2-transfected DCKJJ cells at 7 concentrations from 0.014 μ to 10 μΜ MDCKH cells were maintained in minimal essential medium (MEM) with 1% Pen/Strep, 10% fetal bovine serum, and 0.25 mg mL hygromycin B in an incubator set at 37 °C, 90% humidity and 5% C02. 24 hours prior to assay, media containing 5 mM sodium butyrate were added to MDCKII cells in flasks, and cells were grown to 80-90% confluence. On assay day, cells were trypsinized and resuspended in Krebs-Henseleit Buffer (KHB), pH 7.4 at 5 x 106 million cells/mL. Cells were premcubated for 15 mm in assay plate before addition of test compound or substrate.
Test compounds were serially diluted in DMSO and then spiked (2 μΕ) into in 0.4 mL KHB buffer containing wild-type or OCT2-transfected cells and incubated for 10 minutes. Assay was initiated with the addition of 0.1 mL of 100 μΜ l4C-TEA in KHB buffer (20 μ final concentration after mixing). The concentration of TEA is based on the m. After 10 mmutes of incubation, the assay mixture was quenched with addition of 0.5 mL of ice-cold IX PBS buffer. Samples were then centrifuged at 1000 rpm for 5 min and supernatants were removed. Wash steps were repeated four times with ice-cold PBS. Finally, the cell pellets were lysed with 0.2N NaOH and let sit at room temperature for at least 30 min to ensure complete lysis. Samples were then counted on liquid scintillation counter and dpm counts were used to perform the following calculations. The % inhibition was calculated as follows: % inhibition = [1 - { [OCT2], - [WT]ni } / {[OCr2]3li-[WT]ni} ]* i 00 where, [OCT2], represents the dpm count in the presence of test compound for either OCT2 cells, [OCT2]„j represents the dpm count in the absence of test compound for OCT2 ceils and [WT]ni represents the dpm count in the absence of test compound for wild type cells, respectively.
Table 3
Figure imgf000221_0001
Compound IC50 (nM)
Number
13 610
36 10000
39 358
40 204
41 2823
42 487
45 137
47 6200
48 4909
55 476
63 42
64 94
77 3830
82 10000
83 10000
96 1357
98 3726
99 1506
100 450
The data in Tables 1, 2 and 3 represent an average over time of each assays for each compound. For certain compounds, multiple assays have been conducted over the life of the project. Thus, the data reported in Tables 1, 2 and 3 include the data reported in the priority documents, as well as data from assays run in the intervening period. Example 107
Pharmacokinetic Analysis Following Oral or Intravenous Administration to
Beagle Dogs
Pharmacokinetic analysis was performed on various test compounds following intravenous or oral administration to beagle dogs.
For pharmacokinetic analysis of intravenously administered compounds, the test compounds were formulated in 5% Ethanol, 55% PEG 300, and 40% water at 0.1 mg/mL for IV infusion. For pharmacokinetic analysis of orally administered compounds, the test compounds were formulated as an aqueous suspension in 0.1% Tween 20, 0.5% i f PMC LV100 in Di Water at 1 mg/kg.
Each dosing group consisted of 3 male, non-naive purebred beagle dogs. At dosing, the animals weighed between 10 to 13 kg. The animals were fasted overnight prior to dose administration and up to 4 hr after dosing. For studies of intravenous administration, the test article was administered to the animals by intravenous infusion over 30 min. The rate of infusion was adjusted according to the body weight of each animal to deliver a dose of 0.5 mg/kg. For studies of oral administration, the test article was administered according to the body weight of each animal to deliver a dose of 1 mg/kg.
For pharmacokinetic analysis of intravenously administered compounds, serial venous blood samples (approximately 1 mL each) were taken from each animal at 0, 0.250, 0.483, 0.583, 0.750, 1.00, 1.50, 2.00, 4.00, 8.00, 12.0, and 24.0 hours after dosing. The blood samples were col lected into Vacu tamer1 M tubes containing EDTA- K2 as the anti-coagulant and were immediately placed on wet ice pending centrifugatkm for plasma. An LC MS/MS method was used to measure the concentration of the test compound in plasma. An aliquot of 100 μΕ of each plasma sample was added to a clean 96 well plate, and 400 p.L of cold acetonitrile/internal standard solution (A.CN)/(ISTD) was added. After protein precipitation, an aliquot of 1 10 μΕ of the supernatant was transferred to a clean 96-well plate and diluted with 300 μ.Ε of water. An aliquot of 25 μΐ, of the above solution was injected into a TSQ Quantum Ultra LC/MS/MS system utilizing a Hypersil Gold Cj g HPLC column (50 X 3.0 mm, 5 μιη; Thermo-Hypersil Part # 25105-053030). An Agilent 1200 series binary
??? pump (P/N G1312A Bin Pump) was used for elution and separation, and an HTS Pal autosampler (LEAP Technologies, Carrboro, NC) was used for sample injection, A TSQ Quantum Ultra triple quadrupole mass spectrometer was utilized in selective reaction monitoring mode (Thermo Finnigan, San Jose, CA). Liquid chromatography was performed using two mobile phases: mobile phase A contained 1% acetonitrile in 2.5 mM ammonium formate aqueous solution with pH of 3.0, and mobile phase B contained 90% acetonitrile in 10 mM ammonium formate with pH of 4.6. Non- compartmental pharmacokinetic analysis was performed on the plasma concentration- time data. The resulting data are shown in the first three columns of Table 4. In Table 4, CL refers to clearance, which characterizes the rate at which drug is removed from plasma. The lower the clearance of a drag is, the longer the elimination half-life is in the body. Vss refers to the steady state volume of distribution and indicates how well a drug is distributed into the tissues. The larger the Vss is, the longer the elimination half- life is in the body. MRT refers to mean residence time, which is a measure of the average time molecules exist in the body.
For pharmacokinetic analysis of orally administered compounds, serial venous blood samples (approximately 0.3 mL each) were taken from each animal at time points of 0, 0.25, 0.50, 1.0, 2.0, 4.0, 6.0, 8.0, 12.0 and 24.0 hours after dosing. Blood samples were collected, prepared and analyzed in a similar way to the intranveous studies described above. Non-compartmental pharmacokinetic analysis was performed on the plasma concentration-time data. The resulting data are shown in the last three columns of Table 4. In Table 4, F (%) refers to oral bioavailability. Cmax refers to the peak plasma concentration of the compound after administration. AUC refers to area under the curve and is a measure of total plasma exposure of the indicated compound.
Table 4
Compo CL VM MRT F (%) Cmax (μΜ) AUC
nd # (L/h/kg) (L/kg) (h) aqueous aq eous (uM*h)
suspension suspension aqueous
suspension 98 0.047 0.16 3.3 n/a n/a n/a
83 0.161 0.38 2.4 n/a n/a n a
55 0.058 0.24 4.2 n/a n/a n/a
77 0.300 0.64 2.2 n/a n/a n/a
41 0.015 0.11 7.5 10.7 2.4 16.3
42 0.020 0.15 7.1 28.0 4.5 28.6
47 0.014 0.10 7.4 12.6 2.8 20.4
8 0.498 0.87 1.8 ii-'a n/a n a
7 0.510 1.20 2.3 n/a n a n a
3 0.047 0.23 4.9 18.7 1 ? 9.2
2 0.030 0.20 6.5 40.7 7.8 66.1
All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification are incorporated herein by reference, in their entirety to the extent not inconsistent with the present description.
From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.

Claims

What is claimed is:
A compound having the following Formula (I):
Figure imgf000226_0001
(0 or a stereoisomer or pharmaceutically acceptable salt thereof,
wherein:
X is -O- or -NZ3- or -CHZ3-;
W is -CHZ2-;
ZJ , Z2 and ZJ are each, independently, hydrogen or C1-3alkyl, or wherein 7.} and 7 or Z1 and Z3, taken together, form -L- wherein L is -C(RV, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, or -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, wherein at least one of Z1 and Z2 or 7.} and Zi, taken together, form -L-;
Z4 is a bond, -Ci i,-. or ~CH2CH2~;
Y' and Y"6 are each, independently, hydrogen, C1- alkyl or d-shaloalkyl;
R1 is phenyl substituted with one to three halogens; and
each Ra is, independently, hydrogen, halo, hydroxy! or C.^alkyL
2. A compound of claim 1 having the following Formula ί Π-Λ):
Figure imgf000227_0001
(II-A)
3. A compound of claim 1 having the following Formula (II-B):
Figure imgf000227_0002
(II-B) ompound of claim 1 having the following Formula (II-C)
Figure imgf000227_0003
(II-C ompound of any one of claims 1-4 wherein L is -C(Ra)2-.
6. A compound of any one of claims 1-4 wherein L is
C(Ra)2C(Ra)2-
7. A compound of any one of claims 1-4 wherein L is
-C(R' l)2C(Ra)2C(Ra)2-.
8 A compound of any one of claims 1-7 wherein each Ra is hydrogen.
9. A compound of any one of claims 1-7 wherein one Ra is methyl and eac remaining Ra is hydrogen.
10. A compound of any one of claims 1 -7 wherein at least one Ra is halogen and each remaining Ra is hydrogen.
1 1. A compound of any one of claims 1-7 wherein two Ra are halogen and each remaining Ra is hydrogen.
12. A compound of any one of claims 1-7 wherein one Ra is halogen and each remaining Rd is hydrogen.
13. A compound of any one of claims 1-2 or 5-12 wherein X is -0-.
14. A compound of any one of claims 1-2 or 5-12 wherein X is
NZ3-.
A compound of any one of claims 1-2 or 5-12 wherein X is
-NI-
16. A compound of any one of claims 1-2 or 5-12 wherein X is ■CHZ3- and Z1 and Z3, taken together, form -L-.
17. A compound of claim 16 wherein 7f is hydrogen. A compound of any one of claims 1-2 or 5-12 wherein X is
-CH2-.
A compound of any one of claims 1 or 5-18 wherein Z4 is a bond or -CH,-
20. A compound of any one of claims 1 or 5-18 wherein Z4 is -CH2-
21. A compound of any one of claims 1 or 5-18 wherein Z is a bond.
22. A compound of any one of claims I or 5-21 wherein Y ! and Y2 are each independently hydrogen, methyl or trifluoromethyl.
23. A compound of any one of claims 1-22 wherein R1 is substituted with one halogen.
24. A compound of claim 23 wherein R5 is 4-fl orophenyi or 2- fluorophenyl.
25. A compound of any one of claims 1-22 wherein R1 is substituted with two halogens.
26. A compound of claim 25 wherein R1 is 2,4-difluorophenyL 2,3- di fluorophenyl, 2,6-difluorophenyl, 3-fluoro-4-chlorophenyl, 3,4-difluorophenyl, 2- fluoro-4-chlorophenyl, or 3,5-difluorophenyi.
27. A compound of claim 26 wherein R5 is 2,4-difl.uorophenyl.
28. A compound of any one of claims 1-22 wherein R! is substituted with three halogens.
29. A compound of claim 28 wherein R1 is 2,4,6-irifluorophenyi or,3 ,4-irifluoropheny ί ,
30. A compound of claim 29 wherein R¾ is 2,4,6-trifluorophenyl.
31. A compound of claim 1 selected from :
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000231_0002
Figure imgf000231_0003
Figure imgf000231_0004
Figure imgf000231_0005
230
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000233_0002
Figure imgf000234_0001
Figure imgf000235_0001
234
Figure imgf000236_0001
Figure imgf000237_0001
236
Figure imgf000238_0001
Figure imgf000239_0001
238
Figure imgf000240_0001
239
Figure imgf000241_0001
Figure imgf000241_0002
240
Figure imgf000242_0001
33. A pharmaceutical composition comprising a compound of any one of claims 1-32, or a stereoisomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
34. A pharmaceutical composition of claim 33 further comprising one or more additional therapeutic agents.
35. A pharmaceutical composition of claim 34 wherein the one or more one additional therapeutic agents is an anti-HIV agent.
36. A pharmaceutical composition of claim 35 wherein the one or more additional therapeutic agents is selected from the group consisting of HIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucleotide inhibitors of reverse transcriptase, and combinations thereof.
37. A method of treating an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of claims 1—32 or a pharmaceutical composition of claim 33.
38. A method of claim 37 further comprising administering to the human a therapeutical ly effective amount of one or more additional therapeutic agents.
39. A method of claim 38 wherein the one or more additional therapeutic agents is an anti-HIV agent.
40. A method of claim 39 wherein the one or more additional therapeutic agents is selected from the group consisting of HIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HI V nucleoside inhibitors of reverse transcriptase, HIV nucleotide inhibitors of reverse transcriptase, and combinations thereof.
41. Use of a compound of any one of claims 1-32 or a pharmaceutical composition of claim 33 for the treatment of an HIV infection in a human having or at risk of having the infection.
42. A use of claim 41 further comprising administering to the human a therapeutically effective amount of one or more additional therapeutic agents.
43. A use of claim 42 wherein the one or more additional therapeutic agents is an anti-Hl V agent.
44. A use of claim 43 wherein the one or more additional therapeutic agents is selected from the group consisting of HIV protease mhibitors, HIV non- nucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucleotide inhibitors of reverse transcriptase, and combinations thereof.
45. A compound as described in any one of claims 1-32, or a pharmaceutical ly acceptable salt thereof for use in medical therapy.
46. A compound as described in any one of claims 1-32, or a pharmaceutically acceptable salt thereof, for use in the therapeutic treatment of an HIV infection.
47 A compound having the following Formula (I): Z1 O OH
(I) or a stereoisomer or pharmaceutically acceptable salt thereof,
wherein:
X is -O- or -NZ3- or -CHZ3-;
W is ··()· or -NZ2- or -CHZ2-;
Z1, Z and Z3 are each, independently, hydrogen or C1-3aikyi, or wherem Z1 and Z2 or Z1 and Z3, taken together, form -L- wherein L is ~C(R)2-, ~C(Ra)2C(Ra)2-, ··( (Ra).C( !}>C(ir).- · ( '( R'! }.-( ( W ) ··{ '( R" ) >( 'i R" ) ·- . ··(·( Ry ;,<()( ΪΚ;! ;·.<··.
-C(Ra)2NRaC(R.a)2-, -C(Ra)2SC(Ra)2~, -C(Ra)2S(0)C(Ra)2-, -C(Ra)2S02C(Ra)2-, (YRa)..()(-{R ').<( (R':).'-. -C(Ra)2C(Ra)2OC(Ra)2-, ··( '( R ).·\ R '(. '( R'! ).·( ( \{ ' } · .
-C(Ra)2C(Ra)2NR3C(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -CiRa)2C(Ra)2SC(Ra)2-,
-C(Ra)2S(0)C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2S(0)C(Ra)2-, -C(R )..SC)..C( '} C(R ).-. ··( (Ra).C(R!}>SO>CiR ).-. -C(Ra)2S02Ni aC(Ra)2- or -C(Ra)2NRaS02C(Ra)2-;
Z is a bond or ~CH2-, -Ce2CH2-, -( I !,( !!■( H2-. -Ce2OCH2-, -CH2NRaCH2-, -CH2SCH2- CH2S(0)CH2~ or -CH2S02CH2s
Y1 and Y"6 are each, independently, hydrogen, Chalky! or C1-3haloalkyl, or Y1 and Y% together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms, wherein the carbocyclic or heterocyclic ring is optionally substituted with one or more Ra;
R¾ is optionally substituted aryl or optionally substituted heteroaryi; and each Ra is, independently, hydrogen, halo, hydroxy! or
Figure imgf000244_0001
or wherein two Ra groups, together with the carbon atom to which they are attached, form
OO, and wherein at least one of: (i) Z1 and ' l or Z' and Z3, taken together, form -L-; or (ii) Y1 and Y2, together with the carbon atom to which they are attached, form a carbocvclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
48. A compound of claim 47 wherein W is -€HZ:-
49. A compound of claim 47 or 48 wherein Z! and Z or Z5 and Z' , taken together, form -L-.
Figure imgf000245_0001
Figure imgf000246_0001
wherein L is ••C(Ra),C(K':
-C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Rd)2OC(R , ~C(Ra)2T aC(Ra)2~, ~C(Ra)2SC(Ra)2-, C{Ra)>S(() (! ")·.·. -C(Ra)2S02C(Ra)2-, -C(Ra)2OC(Ra)2C(Ra)2-, -{ ( ;, (-i]r ).ί)(·ίΊΓί -. -C(Ra)2 ]NRaC(Ra)2C,(Ra)2-, -C(R:: ¾ '!)ARi!((R'!)., -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(0)C(R_a)2C(Ra)2~, -C(Ra)2C(Ra)2S(0)C(Ra)2-, -C(Ra)2S02C(Ra)2C(Ra)2-, ··( '( R" ;· >( '( W ) .SO■( '( R" ;·.<·-. -C(Ra)2802NRaC(Ra)2~ or -C(Ra)2NRaS()2C(Ra)2-.
51. A compound of claim 47 or 48 wherein Y and Y together with the carbon atom to which they are attached, form a carbocyclie ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
52. A compound of claim 51 having one of the following Formulas
Figure imgf000246_0002
(III-A)
Figure imgf000247_0001
Figure imgf000247_0002
Figure imgf000247_0003
(III-D)
wherein Z1 and Z3 are each, independently, hydrogen or C1-3alkyl.
53. A compound of claim 51 having one of the following Formulas (III-E), (m-F), (jll-(i) or (III-H):
Figure imgf000248_0001
Figure imgf000248_0002
Figure imgf000248_0003
Figure imgf000248_0004
247 < II I -1 ϊ )
wherein Z1 and 7/ are each, independently, hydrogen or Ci ^alkyl.
54. A compound of claim 47 or 48 wherein both (i) Z and Z or Z and Z3, taken together, form -L-, and (ii) Y1 and Y , together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
55. A compound of claim 54 having one of the following Formulas (IV-AA), (IV-AB), (TV-AC), (IV- AD), (IV- AE), (IV-AF), (1V-AG) or (IV-AH):
Figure imgf000249_0001
Figure imgf000250_0001
Figure imgf000250_0002
Figure imgf000250_0003
(IV-AF)
Figure imgf000251_0001
Figure imgf000251_0002
(IV-AH)
wherein L is
Figure imgf000251_0003
-C(Ra)2C(Ra)2C(Ra)2C(Ra)2- ··( ({ "),()( (R-:),-. -( (R"):NR( (! "),·. -C(Ra)2SC(Ra)2-,
-C(Ra)2S(0)C(Ra)2-, -C(Ra)2S02C(R3)2-, ~c '·: R" ;·.·()( '·: R'! ;·.·( '( W ) >-.
-C(Ra)2C(Ra)2OC(Ra)2-, -(VR ).-\R;('(Ra .-( iR'},-. -αΚ ^Ό^Ι^ θ^)-,
-C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)28C(Ra)2-, -C(Ra)2S(0)C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2S(0)C(Ra)2-, -C(Ra)2S02C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2S02C(Ra)2-,
•C{Ra S().-\ a).<~ or -C(Ra)2NRaS02C(Ra)2-.
56. A compound of claim 54 having one of the following Fonnuias (IV-BA), (IV-BB), (!V-BCk (IV-BD), (IV-BE), (IV-BF), (IV-BG) or (IV-
Figure imgf000252_0001
Figure imgf000252_0002
Figure imgf000252_0003
Figure imgf000252_0004
(IV-BD)
Figure imgf000253_0001
Figure imgf000253_0002
Figure imgf000253_0003
Figure imgf000253_0004
252 (IV-BH)
wherem is
-C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, ··( ( { ") .()( ( R-:) .-. -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(0)C(Ra)2-, -C(Ra)2S02C(Ra)2-, -CiI a)2()Cii 3)2C(Ra)2-,
-C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2NRaC(Ra)2~, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)28C(Ra)2-, -C(Ra)2S(0)C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2S(0)C(Ra)2- -C(Ra)2S02C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2S02C(Ra)2-,
•({R' SO. . a).<~ or -C(Ra)2NRaS02C(Ra)2-.
57. A compound of any one of claims 47-50 or 54-56 wherein L is -C(Ra)2~, -C(Ra)2C(Ra)2-, -C( a)2C(Ra)2C(Ra)2-, or -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-.
58. A compound of claim 57 wherein L is -C(Ra)2-.
59. A compound of claim 57 wherem L is -C(Ra)2C(Ra)2-
60. A compound of claim 57 wherein L is -C(Ra)2C,(Ra)2C(Ra)2-.
61. A compound of any one of claims 47-50 or 54-60 wherem each Ra is hydrogen.
62. A compound of any one of claims 47-50 or 54-60 wherein one Ra is methyl and each remaining R is hydrogen.
63. A compound of any one of claims 47-50 or 54-60 wherein at least one Ra is halogen and each remaining Ra is hydrogen.
64. A compoimd of any one of claims 47-50 or 54-60 wherein two R are halogen and each remaining Ra is hydrogen.
65. A compound of any one of claims 47-50 or 54-60 wherein one RA is halogen and each remaining R is hydrogen.
66. A compound of any one of claims 47-50 or 54-56 wherein L is ··( ( RA )()( ·( R ) . -.
Figure imgf000255_0001
{( :' } >SC( R ! } >-, -C(Ra)28(0)C(RA)2-, or ~C(Ra)2S02C(RA)2~.
67. A compound of any one of claims 47-50 or 54-56 wherein L is -C(RA)2OC(RA)2-.
68. A compound of claim 66 or 67 wherein each RA is hydrogen.
69. A compound of claim 66 or 67 wherein one RA is methyl and eac remaining RA is hydrogen.
70. A compound of claim 66 or 67 wherein at least one R3 is halogen and each remaining R3 is hydrogen,
71. A compound of claim 66 or 67 wherein two RA are halogen and each remaining RA is hydrogen.
72. A compound of claim 66 or 67 wherein one RA is halogen and each remaining RA is hydrogen.
73. A compound of any one of claims 47-55 or 57-72 wherein X is
-0-.
74. A compound of claim 73 wherein Z " is hydrogen.
75. A compound of any one of claims 47-72 wherein X is -NZ -.
76. A compound of any one of claims 47-72 wherein X is -NH-.
77. A compound of any one of claims 47-55 or 57-72 wherem X is
-CHZ3-
78. A compound of any one of claims 47-55 or 57-72 wherem X is -
CH2~,
79. A compound of any one of claims 47-49, 51, 54 or 57-78 wherem Z4 is a bond or -CH2-.
80. A compound of any one of claims 47-49, 51, 54 or 57-78 wherein
Z4 is ~CH2~.
81. A compound of any one of claims 47-49, 51, 54 or 57-78 wherem
Z4 is a bond.
82. A compound of any one of claims 47-49 or 57-81 wherem Y1 and Y2 are each independently hydrogen, methyl or trifluoromethyl.
83. A compound of any one of claims 47-82 wherein R! is phenyl.
84. A compound of any one of claims 47-82 wherem R1 is pyridinyl.
85. A compound of any one of claims 47-84 wherein R1 is substituted with at least one halogen.
86. A compound of any one of claims 47-84 wherein R1 is substituted with one halogen.
87. A compound of claim 86 wherein R1 is 4-fluorophenyl or 2~ fluorophenyl.
88. A compound of any one of claims 47-84 wherein R1 is substituted with two halogens,
89. A compound of claim 88 wherein R! is 2,4-difliiorophenyl, 2,3- difiuorophenyl, 2,6-difluorophenyL 3-fluoro-4-chloropheiiyl, 3,4-difluoropheiiyl, 2- ftuoro-4-chlorophenyl, or 3,5-difluorophenyl.
90. A compound of claim 89 wherein R1 is 2,4-difl.uorophenyl.
91 . A compound of any one of claims 47-84 wherein R] is substituted with three halogens.
92. A compound of claim 91 wherein R1 is 2,4,6-trifluorophenyl or 2,3,4-trifluoiOphenyi,
93. A compound of claim 92 wherein R3 is 2,4,6-trifluorophenyl .
94. A compound of claim 85 wherein R1 is 3-trifluoromethyl-4- fluorophenyi or 2-cyclopropoxy-4-fluorophenyl.
95. A pharmaceutical composition comprising a compound of any one of claims 47-94, or a stereoisomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable earner, diluent or excipient.
96. A pharmaceutical composition of claim 95 further comprising one or more additional therapeutic agents.
97. A phannaceutical composition of claim 96 wherein the one or more one additional therapeutic agents is an anti-HIV agent.
98. A pharmaceutical composition of claim 97 wherein the one or more additional therapeutic agents is selected from the group consisting of HIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HIV nucleoside mhibitors of reverse transcriptase, HIV nucleotide inhibitors of reverse transcriptase, and combinations thereof.
99. A method of treating an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of claims 47-94 or a pharmaceutical composition of claim 95.
100. A method of claim 99 further comprising administering to the human a therapeutically effective amount of one or more additional therapeutic agents.
101. A method of claim 100 wherein the one or more additional therapeutic agents is an anti-HIV agent.
102. A method of claim 101 wherein the one or more additional therapeutic agents is selected from the group consisting of HIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HI V nucleoside inhibitors of reverse transcriptase, HIV nucleotide mhibitors of reverse transcriptase, and other drugs for treating HIV, and combinations thereof.
103. Use of a compound of any one of claims 47-94 or a pharmaceutical composition of claim 95 for the treatment of an HIV infection in a human having or at risk of having the infection.
104. A use of claim 103 further comprising administering to the human a therapeutically effective amount of one or more additional therapeutic agents.
105. A use of claim 104 wherein the one or more additional therapeutic agents is an anti-HIV agent.
106. A use of claim 105 wherein the one or more additional therapeutic agents is selected from the group consisting of HIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucleotide mhibitors of reverse transcriptase, and other drugs for treating HIV, and combinations thereof.
107. A compound as described in any one of claims 47-94, or a pharmaceutically acceptable salt thereof for use in medical therapy.
108. A compound as described in any one of claims 47-94, or a pharmaceutically acceptable salt thereof, for use in the therapeutic treatment of an HIV infection.
PCT/US2013/076367 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use WO2014100323A1 (en)

Priority Applications (60)

Application Number Priority Date Filing Date Title
KR1020237014240A KR20230061576A (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
KR1020157019194A KR101770048B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
PH12019501848A PH12019501848A1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
BR122015029881-4A BR122015029881B1 (en) 2012-12-21 2013-12-19 Polycyclic carbamoylpyridone compounds, pharmaceutical composition comprising them and their pharmaceutical use
KR1020217012463A KR102351912B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
NZ709260A NZ709260A (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
KR1020207015960A KR102246963B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
DK13815937.1T DK2822954T3 (en) 2012-12-21 2013-12-19 POLYCYCLIC-CARBAMOYLPYRIDONE COMPOUNDS AND THEIR PHARMACEUTICAL USE
EA201591027A EA030003B1 (en) 2012-12-21 2013-12-19 Polycyclic carbamoylpyridone compound and pharmaceutical use thereof for treating hiv infection
EP22178867.2A EP4122935A1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
UAA201506209A UA114351C2 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
EP15201519.4A EP3067358B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
MX2017000362A MX357940B (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use.
CN201380073134.XA CN104995198B (en) 2012-12-21 2013-12-19 Polycyclic carbamoylpyridone compound and its medicinal usage
MDA20200093A MD4841B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
MEP-2016-93A ME02400B (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
MDA20170067A MD4754C1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
KR1020227018650A KR102527797B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
CA2893843A CA2893843C (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
RS20160407A RS54873B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
BR112015014714-3A BR112015014714B1 (en) 2012-12-21 2013-12-19 polycyclic carbamoylpyridone compounds, pharmaceutical compositions comprising them and their pharmaceutical use
JP2015549669A JP6028105B2 (en) 2012-12-21 2013-12-19 Polycyclic carbamoylpyridone compound and pharmaceutical use thereof
CR20170279A CR20170279A (en) 2012-12-21 2013-12-19 POLYCHYCLIC CARBAMOILPIRIDONE COMPOUNDS AND THEIR PHARMACEUTICAL USE
KR1020177016697A KR102040007B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
SI201330197A SI2822954T1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
CN202310322212.0A CN116640140A (en) 2012-12-21 2013-12-19 Polycyclic-carbamoyl pyridone compounds and pharmaceutical uses thereof
MDA20150064A MD4736C1 (en) 2012-12-21 2013-12-19 Polycyclic carbamoylpyridone compound and its pharmaceutical use
KR1020197031864A KR102120875B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
KR1020227001078A KR102406288B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
ES13815937.1T ES2577283T3 (en) 2012-12-21 2013-12-19 Polycyclic carbamoyl-pyridone compounds and their pharmaceutical use
PL15201519T PL3067358T3 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
CN202010188107.9A CN111303152B (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
EP13815937.1A EP2822954B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
MX2015008009A MX344879B (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use.
EP19185121.1A EP3608325B1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
SG11201504857SA SG11201504857SA (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
AU2013361401A AU2013361401C1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
IL239316A IL239316A (en) 2012-12-21 2015-06-10 Polycylic-carbamoylpyridone compounds and their pharmaceutical use
PH12015501445A PH12015501445B1 (en) 2012-12-21 2015-06-22 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
HK15105909.2A HK1205124A1 (en) 2012-12-21 2015-06-22 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
CR20150380A CR20150380A (en) 2012-12-21 2015-07-17 POLYCHYCLIC CARBAMOILPIRIDONE COMPOUNDS AND THEIR PHARMACEUTICAL USE
ZA2015/07997A ZA201507997B (en) 2012-12-21 2015-10-26 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
PH12016500389A PH12016500389B1 (en) 2012-12-21 2016-02-29 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
HK16104629.3A HK1216643A1 (en) 2012-12-21 2016-04-21 Polycyclic carbamoylpyridone compounds and their pharmaceutical use
HRP20160544TT HRP20160544T1 (en) 2012-12-21 2016-05-20 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
IL245780A IL245780B (en) 2012-12-21 2016-05-23 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
SM201600157T SMT201600157B (en) 2012-12-21 2016-06-01 POLYCYCLIC CARBAMMYL PYRIDONE COMPOUNDS AND THEIR PHARMACEUTICAL USE
IL247311A IL247311A0 (en) 2012-12-21 2016-08-17 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
AU2016262722A AU2016262722B2 (en) 2012-12-21 2016-11-24 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
LU00083C LUC00083I2 (en) 2012-12-21 2018-08-17 Bictegravir or a pharmaceutically acceptable salt thereof, in particular bictegravir sodium
LTPA2018511C LTC2822954I2 (en) 2012-12-21 2018-08-21 Polycyclic carbamoylpyridine compounds and their pharmaceutical applications
NL300947C NL300947I2 (en) 2012-12-21 2018-08-22 Bictegravir or a pharmaceutically acceptable salt thereof, in particular bictegravir sodium
CY2018022C CY2018022I2 (en) 2012-12-21 2018-08-23 POLYCYCLIC CARBAMOYLPYRIDONE COMPOUNDS AND THEIR PHARMACEUTICAL USE
HUS1800035C HUS1800035I1 (en) 2012-12-21 2018-08-23 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
AU2018232957A AU2018232957B2 (en) 2012-12-21 2018-09-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
CY20191101151T CY1122246T1 (en) 2012-12-21 2019-11-05 POLYCYCLIC CARBAMOYLPYRIDONE COMPOUNDS AND THEIR PHARMACEUTICAL USE
AU2020202914A AU2020202914B2 (en) 2012-12-21 2020-05-01 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
IL274988A IL274988A (en) 2012-12-21 2020-05-28 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
AU2021221427A AU2021221427B2 (en) 2012-12-21 2021-08-23 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
AU2023286038A AU2023286038A1 (en) 2012-12-21 2023-12-30 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201261745375P 2012-12-21 2012-12-21
US61/745,375 2012-12-21
US201361788397P 2013-03-15 2013-03-15
US61/788,397 2013-03-15
US201361845803P 2013-07-12 2013-07-12
US61/845,803 2013-07-12

Publications (1)

Publication Number Publication Date
WO2014100323A1 true WO2014100323A1 (en) 2014-06-26

Family

ID=49917297

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/076367 WO2014100323A1 (en) 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use

Country Status (41)

Country Link
US (6) US9216996B2 (en)
EP (4) EP2822954B1 (en)
JP (6) JP6028105B2 (en)
KR (8) KR102527797B1 (en)
CN (4) CN111303152B (en)
AP (1) AP2015008510A0 (en)
AR (1) AR094197A1 (en)
AU (6) AU2013361401C1 (en)
BR (2) BR112015014714B1 (en)
CA (3) CA3012242C (en)
CL (3) CL2015001756A1 (en)
CR (2) CR20170279A (en)
CY (3) CY1117570T1 (en)
DK (3) DK3067358T3 (en)
EA (2) EA037633B1 (en)
ES (3) ES2753548T3 (en)
HK (3) HK1205124A1 (en)
HR (3) HRP20220899T1 (en)
HU (4) HUE046559T2 (en)
IL (4) IL239316A (en)
LT (3) LT3067358T (en)
LU (1) LUC00083I2 (en)
MD (3) MD4754C1 (en)
ME (1) ME02400B (en)
MX (4) MX344879B (en)
MY (2) MY164352A (en)
NL (1) NL300947I2 (en)
NZ (2) NZ709260A (en)
PE (2) PE20170528A1 (en)
PH (3) PH12019501848A1 (en)
PL (3) PL3067358T3 (en)
PT (3) PT2822954E (en)
RS (1) RS54873B1 (en)
SG (1) SG11201504857SA (en)
SI (3) SI2822954T1 (en)
SM (1) SMT201600157B (en)
TW (5) TWI693224B (en)
UA (2) UA123572C2 (en)
UY (1) UY35213A (en)
WO (1) WO2014100323A1 (en)
ZA (2) ZA201507997B (en)

Cited By (131)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015006731A1 (en) * 2013-07-12 2015-01-15 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their use for the treatment of hiv infections
WO2015006733A1 (en) * 2013-07-12 2015-01-15 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their use for the treatment of hiv infections
WO2015177537A1 (en) * 2014-05-20 2015-11-26 Cipla Limited Process for preparing polycyclic carbamoyl pyridone derivatives and intermediates thereof
US9216996B2 (en) 2012-12-21 2015-12-22 Gilead Sciences, Inc. Substituted 2,3,4,5,7,9,13,13a-octahydropyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazepines and methods for treating viral infections
WO2015196137A1 (en) * 2014-06-20 2015-12-23 Gilead Sciences, Inc. Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5] pyrazino [2,1-b] [1,3] oxazepine-10-carboxamide
WO2015196116A1 (en) * 2014-06-20 2015-12-23 Gilead Sciences, Inc. Sodium (2r, 5s, 13ar) -7, 9-dioxo-10- ( (2,4,6-trifluorobenzyl) carbamoyl) -2, 3, 4, 5, 7, 9, 13, 13a-octahydro-2, 5-methanopyrido [1',2' : 4.5] pyrazino [2, 1-b] oxazepin-8-olate
WO2016027879A1 (en) * 2014-08-22 2016-02-25 塩野義製薬株式会社 Polycyclic pyridone derivative having integrase-inhibiting activity
WO2016036759A1 (en) 2014-09-04 2016-03-10 Gilead Sciences, Inc. Methods of treating or preventing hiv in patients using a combination of tenofovir alafenamide and dolutegravir
WO2015195656A3 (en) * 2014-06-20 2016-03-24 Gilead Sciences ,Inc. Synthesis of polycyclic-carbamoylpyridone compounds
WO2016105564A1 (en) 2014-12-24 2016-06-30 Gilead Sciences, Inc. Quinazoline derivatives used to treat hiv
WO2016105534A1 (en) 2014-12-24 2016-06-30 Gilead Sciences, Inc. Isoquinoline compounds for the treatment of hiv
WO2016105532A1 (en) 2014-12-24 2016-06-30 Gilead Sciences, Inc. Fused pyrimidine compounds for the treatment of hiv
WO2016106237A1 (en) 2014-12-23 2016-06-30 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
WO2016141092A1 (en) 2015-03-04 2016-09-09 Gilead Sciences, Inc. Toll-like receptor modulating 4,6-diamino-pyrido[3,2-d]pyrimidine compounds
WO2016161382A1 (en) 2015-04-02 2016-10-06 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
WO2017035230A1 (en) 2015-08-26 2017-03-02 Gilead Sciences, Inc. Deuterated toll-like receptor modulators
WO2017059120A1 (en) 2015-09-30 2017-04-06 Gilead Sciences, Inc. Compounds and combinations for the treatment of hiv
WO2017083304A1 (en) 2015-11-09 2017-05-18 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
WO2017106346A2 (en) 2015-12-15 2017-06-22 Gilead Sciences, Inc. Human immunodeficiency virus neutralizing antibodies
US9688695B2 (en) 2012-04-26 2017-06-27 Bristol-Myers Squibb Company Imidazothiadiazole and imidazopyrazine derivatives as protease activated receptor 4 (PAR4) inhibitors for treating platelet aggregation
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
WO2018035359A1 (en) 2016-08-19 2018-02-22 Gilead Sciences, Inc. Therapeutic compounds useful for the prophylactic or therapeutic treatment of an hiv virus infection
WO2018042331A1 (en) 2016-08-31 2018-03-08 Glaxosmithkline Intellectual Property (No.2) Limited Combinations and uses and treatments thereof
WO2018042332A1 (en) 2016-08-31 2018-03-08 Glaxosmithkline Intellectual Property (No.2) Limited Combinations and uses and treatments thereof
WO2018045150A1 (en) 2016-09-02 2018-03-08 Gilead Sciences, Inc. 4,6-diamino-pyrido[3,2-d]pyrimidine derivaties as toll like receptor modulators
WO2018045144A1 (en) 2016-09-02 2018-03-08 Gilead Sciences, Inc. Toll like receptor modulator compounds
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
WO2018051250A1 (en) 2016-09-14 2018-03-22 Viiv Healthcare Company Combination comprising tenofovir alafenamide, bictegravir and 3tc
WO2018064080A1 (en) 2016-09-28 2018-04-05 Gilead Sciences, Inc. Benzothiazol-6-yl acetic acid derivatives and their use for treating hiv infection
WO2018064071A1 (en) 2016-09-27 2018-04-05 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
US9949955B2 (en) 2012-06-15 2018-04-24 Glenmark Pharmaceuticals S.A. Triazolone compounds as mPGES-1 inhibitors
WO2018081292A1 (en) 2016-10-27 2018-05-03 Gilead Sciences, Inc. Crystalline forms of darunavir free base, hydrate, solvates and salts
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
WO2018144390A1 (en) 2017-01-31 2018-08-09 Gilead Sciences, Inc. Crystalline forms of tenofovir alafenamide
WO2018145021A1 (en) 2017-02-06 2018-08-09 Gilead Sciences, Inc. Atazanavir (atv) analogues for treating hiv infections
WO2018229798A1 (en) 2017-06-13 2018-12-20 Cipla Limited Process for the preparation of bictegravir and intermediate thereof
WO2018237148A1 (en) 2017-06-21 2018-12-27 Gilead Sciences, Inc. Multispecific antibodies that target hiv gp120 and cd3
WO2019027920A1 (en) 2017-08-01 2019-02-07 Gilead Sciences, Inc. Crystalline forms of ethyl ((s)-((((2r,5r)-5-(6-amino-9h-purin-9-yl)-4-fluoro-2,5-dihydrofuran-2-yl)oxy)methyl)(phenoxy)phosphoryl)-l-alaninate (gs-9131) for treating viral infections
WO2019035904A1 (en) 2017-08-17 2019-02-21 Gilead Sciences, Inc. Solid forms of an hiv capsid inhibitor
WO2019035973A1 (en) 2017-08-17 2019-02-21 Gilead Sciences, Inc Choline salt forms of an hiv capsid inhibitor
US10246433B2 (en) 2012-12-21 2019-04-02 Pfizer Inc. Aryl and heteroaryl fused lactams
WO2019075291A1 (en) 2017-10-13 2019-04-18 Gilead Sciences, Inc. 1-benzyl-2-imino-4-phenyl-5-oxoimidazolidine derivatives as hiv protease inhibitors
WO2019084020A1 (en) 2017-10-24 2019-05-02 Gilead Sciences, Inc. Methods of treating patients co-infected with a virus and tuberculosis
WO2019113462A1 (en) 2017-12-07 2019-06-13 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2019154634A1 (en) 2018-02-09 2019-08-15 Sandoz Ag Crystalline form of bictegravir sodium
WO2019159199A1 (en) 2018-02-16 2019-08-22 Cipla Limited Continues flow process for the preparation of active pharmaceutical ingredients - polycyclic carbamoyl pyridone derivatives and intermediates thereof
WO2019161017A1 (en) 2018-02-15 2019-08-22 Gilead Sciences, Inc. Pyridine derivatives and their use for treating hiv infection
WO2019193542A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides
WO2019193533A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'2'-cyclic dinucleotides
WO2019193543A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
WO2019211799A1 (en) 2018-05-03 2019-11-07 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide
US10479801B2 (en) 2016-12-02 2019-11-19 Merck Sharp & Dohme Corp. Tetracyclic heterocycle compounds useful as HIV integrase inhibitors
WO2019230858A1 (en) 2018-05-31 2019-12-05 塩野義製薬株式会社 Polycyclic carbamoylpyridone derivative
WO2020003151A1 (en) * 2018-06-28 2020-01-02 Honour Lab Limited Process for the preparation of sodium (2r,5s,13ar)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1', 2':4,5]pyrazino[2,1-b] [1,3] oxazepin-8-olate and its polymorphic form
WO2020004443A1 (en) 2018-06-27 2020-01-02 国立大学法人北海道大学 Arenavirus growth inhibitor comprising polycyclic carbamoyl pyridone derivative
WO2020010107A1 (en) 2018-07-03 2020-01-09 Gilead Sciences, Inc. Antibodies that target hiv gp120 and methods of use
WO2020018459A1 (en) 2018-07-16 2020-01-23 Gilead Sciences, Inc. Capsid inhibitors for the treatment of hiv
RU2712275C2 (en) * 2015-04-28 2020-01-28 Сионоги Энд Ко., Лтд. Substituted polycyclic pyridone derivatives and prodrugs thereof
WO2020028272A1 (en) 2018-07-30 2020-02-06 Gilead Sciences, Inc. Anti-hiv compounds
US10570121B2 (en) 2014-06-17 2020-02-25 Pfizer Inc. Substituted dihydroisoquinolinone compounds
WO2020012408A3 (en) * 2018-07-12 2020-03-05 Laurus Labs Limited A process for purification of protected polycyclic carbamoylpyridone derivatives
RU2717101C1 (en) * 2019-06-03 2020-03-18 Андрей Александрович Иващенко Anellated 9-hydroxy-1,8-dioxo-1,3,4,8-tetrahydro-2h-pyrido[1,2-a]pyrazine-7-carboxamides - integrase inhibitors, methods for preparing and using thereof
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
WO2020072656A1 (en) 2018-10-03 2020-04-09 Gilead Sciences, Inc. Imidozopyrimidine derivatives
WO2020092528A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity
WO2020092621A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
EP3653629A1 (en) 2018-11-16 2020-05-20 Sandoz AG Acid addition salts of an integrase strand transfer inhibitor
EP3664896A1 (en) 2017-08-09 2020-06-17 VIIV Healthcare Company Combinations and uses and treatments thereof
EP3664895A1 (en) 2017-08-09 2020-06-17 VIIV Healthcare Company Combinations and uses and treatments thereof
US10696657B2 (en) 2018-02-16 2020-06-30 Gilead Sciences, Inc. Methods and intermediates for preparing therapeutic compounds
WO2020161744A1 (en) 2019-02-07 2020-08-13 Cipla Limited Novel polymorphs of integrase inhibitor
WO2020176505A1 (en) 2019-02-25 2020-09-03 Gilead Sciences, Inc. Protein kinase c agonists
WO2020176510A1 (en) 2019-02-25 2020-09-03 Gilead Sciences, Inc. Protein kinase c agonists
WO2020178770A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides and prodrugs thereof
WO2020178769A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides and prodrugs thereof
WO2020178768A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
WO2020197991A1 (en) 2019-03-22 2020-10-01 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and their pharmaceutical use
WO2020214663A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020214652A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020214647A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of an hiv protease inhibitor
WO2020214716A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. 2-imino-5-oxo-imidazolidine inhibitors of hiv protease
WO2020236753A1 (en) 2019-05-21 2020-11-26 Gilead Sciences, Inc. Methods of identifying hiv patients sensitive to therapy with gp120 v3 glycan-directed antibodies
WO2020237025A1 (en) 2019-05-23 2020-11-26 Gilead Sciences, Inc. Substituted exo-methylene-oxindoles which are hpk1/map4k1 inhibitors
WO2020255038A1 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company Combination of hepatitis b virus (hbv) vaccines and pyridopyrimidine derivatives
WO2020263830A1 (en) 2019-06-25 2020-12-30 Gilead Sciences, Inc. Flt3l-fc fusion proteins and methods of use
WO2021011891A1 (en) 2019-07-18 2021-01-21 Gilead Sciences, Inc. Long-acting formulations of tenofovir alafenamide
WO2021011544A1 (en) 2019-07-16 2021-01-21 Gilead Sciences, Inc. Hiv vaccines and methods of making and using
WO2021034804A1 (en) 2019-08-19 2021-02-25 Gilead Sciences, Inc. Pharmaceutical formulations of tenofovir alafenamide
US10966999B2 (en) 2017-12-20 2021-04-06 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2021107066A1 (en) 2019-11-28 2021-06-03 塩野義製薬株式会社 Prophylactic and therapeutic pharmaceutical agent for hiv infectious diseases characterized by comprising combination of integrase inhibitor and anti-hiv agent
US11034668B2 (en) 2011-07-06 2021-06-15 Gilead Sciences, Inc. Compounds for the treatment of HIV
WO2021130638A1 (en) 2019-12-24 2021-07-01 Carna Biosciences, Inc. Diacylglycerol kinase modulating compounds
WO2021173522A1 (en) 2020-02-24 2021-09-02 Gilead Sciences, Inc. Tetracyclic compounds for treating hiv infection
WO2021188959A1 (en) 2020-03-20 2021-09-23 Gilead Sciences, Inc. Prodrugs of 4'-c-substituted-2-halo-2'-deoxyadenosine nucleosides and methods of making and using the same
EP3693373A4 (en) * 2017-10-06 2021-11-03 Shionogi & Co., Ltd. Method for stereoselectively producing substituted polycyclic pyridone derivative
WO2021236944A1 (en) 2020-05-21 2021-11-25 Gilead Sciences, Inc. Pharmaceutical compositions comprising bictegravir
CN113698420A (en) * 2020-05-22 2021-11-26 上海迪赛诺生物医药有限公司 Novel crystal form of bicalutavir sodium and preparation method thereof
US11203610B2 (en) 2017-12-20 2021-12-21 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2021262990A1 (en) 2020-06-25 2021-12-30 Gilead Sciences, Inc. Capsid inhibitors for the treatment of hiv
WO2022031894A1 (en) 2020-08-07 2022-02-10 Gilead Sciences, Inc. Prodrugs of phosphonamide nucleotide analogues and their pharmaceutical use
US11248005B2 (en) 2019-07-08 2022-02-15 Lupin Limited Process for preparation of intermediates used for the synthesis of HIV integrase inhibitor
US11253524B2 (en) 2018-01-19 2022-02-22 Gilead Sciences, Inc. Metabolites of bictegravir
WO2022072520A1 (en) 2020-09-30 2022-04-07 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and uses thereof
WO2022087149A2 (en) 2020-10-22 2022-04-28 Gilead Sciences, Inc. Interleukin-2-fc fusion proteins and methods of use
CN114426540A (en) * 2020-10-29 2022-05-03 上海拓界生物医药科技有限公司 Pyrido [1,2-a ] pyrazine-1, 8-diketone prodrug derivatives, preparation method and application thereof
WO2022103758A1 (en) 2020-11-11 2022-05-19 Gilead Sciences, Inc. METHODS OF IDENTIFYING HIV PATIENTS SENSITIVE TO THERAPY WITH gp120 CD4 BINDING SITE-DIRECTED ANTIBODIES
US11358711B2 (en) 2019-06-20 2022-06-14 Merck Sharp & Dohme Corp. Tetracyclic heterocycle compounds useful as HIV integrase inhibitors
WO2022159387A1 (en) 2021-01-19 2022-07-28 Gilead Sciences, Inc. Substituted pyridotriazine compounds and uses thereof
US11453669B2 (en) 2018-05-31 2022-09-27 Shionogi & Co., Ltd. Polycyclic pyridone derivative
WO2022245671A1 (en) 2021-05-18 2022-11-24 Gilead Sciences, Inc. Methods of using flt3l-fc fusion proteins
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2023102529A1 (en) 2021-12-03 2023-06-08 Gilead Sciences, Inc. Therapeutic compounds for hiv virus infection
WO2023102239A1 (en) 2021-12-03 2023-06-08 Gilead Sciences, Inc. Therapeutic compounds for hiv virus infection
WO2023102523A1 (en) 2021-12-03 2023-06-08 Gilead Sciences, Inc. Therapeutic compounds for hiv virus infection
WO2023196875A1 (en) 2022-04-06 2023-10-12 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and uses thereof
EP3993797A4 (en) * 2019-06-18 2023-10-25 Laurus Labs Limited Process and polymorphic forms of bictegravir and its pharmaceutically acceptable salts or co-crystals thereof
US11807625B2 (en) 2019-11-26 2023-11-07 Gilead Sciences, Inc. Capsid inhibitors for the prevention of HIV
WO2024006982A1 (en) 2022-07-01 2024-01-04 Gilead Sciences, Inc. Therapeutic compounds useful for the prophylactic or therapeutic treatment of an hiv virus infection
WO2024015741A1 (en) 2022-07-12 2024-01-18 Gilead Sciences, Inc. Hiv immunogenic polypeptides and vaccines and uses thereof
WO2024044477A1 (en) 2022-08-26 2024-02-29 Gilead Sciences, Inc. Dosing and scheduling regimen for broadly neutralizing antibodies
WO2024076915A1 (en) 2022-10-04 2024-04-11 Gilead Sciences, Inc. 4'-thionucleoside analogues and their pharmaceutical use
US12024528B2 (en) 2023-06-14 2024-07-02 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and uses thereof

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016090545A1 (en) 2014-12-09 2016-06-16 Merck Sharp & Dohme Corp. Spirocyclic heterocycle compounds useful as hiv integrate inhibitors
WO2017197055A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Heterocyclic degronimers for target protein degradation
WO2017197046A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. C3-carbon linked glutarimide degronimers for target protein degradation
CN109562113A (en) 2016-05-10 2019-04-02 C4医药公司 Loop coil degron body for target protein degradation
WO2018005328A1 (en) * 2016-06-27 2018-01-04 Concert Pharmaceuticals, Inc. Deuterated bictegravir
CN108250215B (en) * 2016-12-28 2022-04-19 华创合成制药股份有限公司 Novel anti-HIV medicine, preparation method and application thereof
CN106860464A (en) * 2017-02-16 2017-06-20 江苏艾迪药业有限公司 Pharmaceutical composition and application for combination antiviral therapy
CN106860414B (en) * 2017-02-16 2019-12-24 江苏艾迪药业股份有限公司 anti-HIV compound preparation and preparation method and application thereof
CN109020911B (en) * 2018-04-16 2022-06-28 常州制药厂有限公司 Intermediate for preparing bictegravir and preparation method thereof
US11466031B2 (en) 2018-04-26 2022-10-11 Mylan Laboratories Limited Polymorphic forms of bictegravir and its sodium salt
CN110526930B (en) * 2018-05-23 2022-06-03 莫云芬 anti-HIV (human immunodeficiency virus) sulfur-containing polycyclic-hydroxypyridone formamide analogue and application thereof
CN112996517A (en) 2018-09-19 2021-06-18 吉利德科学公司 Integrase inhibitors for the prevention of HIV
CN113226319B (en) 2018-10-22 2023-04-11 内布拉斯加大学董事会 Antiviral prodrugs and nanoformulations thereof
CN110229174A (en) * 2019-05-22 2019-09-13 博诺康源(北京)药业科技有限公司 The synthetic method of Bictegravir bulk pharmaceutical chemicals genotoxicity impurity
CN110263404B (en) * 2019-06-12 2023-03-21 江苏大学 Method for calculating deposition characteristics of integrated prefabricated pump station based on DPM model
CN110698473B (en) * 2019-10-08 2020-12-18 浙江大学 Piperazinone-hydroxypyridone-5-carboxyl compound, preparation and application
JPWO2021107065A1 (en) * 2019-11-28 2021-06-03
WO2022089562A1 (en) * 2020-10-30 2022-05-05 上海拓界生物医药科技有限公司 Use of inhibiting genetically defective hiv virus
WO2023006087A1 (en) * 2021-07-30 2023-02-02 南京明德新药研发有限公司 Macrocyclic pyridone compound and application thereof
CN114230579A (en) * 2021-11-12 2022-03-25 南京艾迪医药科技有限公司 Polycyclic carbamoylpyridone derivatives, process for their preparation and pharmaceutical compositions containing them
CN114605437A (en) * 2022-04-01 2022-06-10 遵义医科大学 Synthesis process for preparing three telaprevir medicaments by continuous one-pot method
WO2023248240A1 (en) 2022-06-21 2023-12-28 Mylan Laboratories Limited Polymorphic forms of bictegravir sodium
WO2024061257A1 (en) * 2022-09-20 2024-03-28 吉斯凯(苏州)制药有限公司 Polycyclic n-heterocyclic ketone compound and use thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006116764A1 (en) * 2005-04-28 2006-11-02 Smithkline Beecham Corporation Polycyclic carbamoylpyridone derivative having hiv integrase inhibitory activity
EP2412709A1 (en) * 2009-03-26 2012-02-01 Shionogi & Co., Ltd. Process for producing pyrone and pyridone derivatives

Family Cites Families (132)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE788516A (en) 1971-09-10 1973-03-07 Lonza Ag METHOD OF MANUFACTURING ALCOXYACETYLACETIC ESTERS
GB1528382A (en) 1974-12-26 1978-10-11 Teijin Ltd Cyclopentene diols and acyl esters thereof and processes for their preparation
DE2658401A1 (en) 1976-12-23 1978-07-06 Merck Patent Gmbh CYCLOPENTAN-1-AMINE, METHOD FOR THE PRODUCTION THEREOF AND AGENTS CONTAINING THESE COMPOUNDS
US4575694A (en) 1984-03-05 1986-03-11 Allied Corporation Coaxial connector
DE3900736A1 (en) 1989-01-12 1990-07-26 Hoechst Ag POSITIVELY WORKING RADIATION-SENSITIVE MIXTURE CONTAINING A MULTI-FUNCTIONAL (ALPHA) -DIAZO- (BETA) -KETOESTER, METHOD FOR THE PRODUCTION THEREOF AND RADIATION-SENSITIVE RECORDING MATERIAL COMPRISING THIS MIXTURE
US5204466A (en) 1990-02-01 1993-04-20 Emory University Method and compositions for the synthesis of bch-189 and related compounds
US6703396B1 (en) 1990-02-01 2004-03-09 Emory University Method of resolution and antiviral activity of 1,3-oxathiolane nuclesoside enantiomers
US5914331A (en) 1990-02-01 1999-06-22 Emory University Antiviral activity and resolution of 2-hydroxymethyl-5-(5-fluorocytosin-1-yl)-1,3-oxathiolane
DE4014649A1 (en) 1990-05-08 1991-11-14 Hoechst Ag NEW MULTIFUNCTIONAL CONNECTIONS WITH (ALPHA) -DIAZO-SS-KETOESTER AND SULPHONIC ACID UNIT UNITS, METHOD FOR THEIR PRODUCTION AND USE THEREOF
GB9301000D0 (en) 1993-01-20 1993-03-10 Glaxo Group Ltd Chemical compounds
US5922695A (en) 1996-07-26 1999-07-13 Gilead Sciences, Inc. Antiviral phosphonomethyoxy nucleotide analogs having increased oral bioavarilability
SE9702772D0 (en) 1997-07-22 1997-07-22 Astra Pharma Prod Novel compounds
US5935946A (en) 1997-07-25 1999-08-10 Gilead Sciences, Inc. Nucleotide analog composition and synthesis method
AU1403099A (en) 1997-11-14 1999-06-07 Merck & Co., Inc. Alpha-1a adrenergic receptor antagonists
JP2002529455A (en) 1998-11-09 2002-09-10 ジェームズ・ブラック・ファウンデーション・リミテッド Gastrin and cholecystokinin receptor ligands
GB2345058A (en) 1998-12-01 2000-06-28 Cerebrus Pharm Ltd Hydroxypyridone compounds useful in the treatment of oxidative damage to the central nervous system
DE69939749D1 (en) 1998-12-25 2008-11-27 Shionogi & Co AROMATIC HETEROCYCLES WITH HIV INTEGRASE INHIBITING PROPERTIES
AU2001262732A1 (en) 2000-06-14 2001-12-24 Shionogi And Co., Ltd. Inhibitor for enzyme having two divalent metal ions as active centers
WO2002030426A1 (en) * 2000-10-12 2002-04-18 Merck & Co., Inc. Aza- and polyaza-naphthalenyl-carboxamides useful as hiv integrase inhibitors
CN101513402B (en) 2001-08-10 2012-03-21 盐野义制药株式会社 Antiviral agent
MY169670A (en) 2003-09-03 2019-05-08 Tibotec Pharm Ltd Combinations of a pyrimidine containing nnrti with rt inhibitors
IL160642A0 (en) 2001-10-03 2004-07-25 Ucb Sa Pyrrolidinone derivatives
BRPI0213522C1 (en) 2001-10-26 2021-05-25 St Di Ricerche Di Biologia Molecolare P Angeletti S P A hydroxypyrimidinone derivative compounds, pharmaceutical composition, and use of a compound
US7109186B2 (en) 2002-07-09 2006-09-19 Bristol-Myers Squibb Company HIV integrase inhibitors
AU2003267098B2 (en) 2002-09-11 2008-11-20 Merck & Co., Inc. Dihydroxypyridopyrazine-1,6-dione compounds useful as HIV integrase inhibitors
EP4059923A1 (en) 2002-11-20 2022-09-21 Japan Tobacco Inc. 4-oxoquinoline compound and use thereof as hiv integrase inhibitor
US20040224917A1 (en) 2003-01-14 2004-11-11 Gilead Sciences, Inc. Compositions and methods for combination antiviral therapy
TW200510425A (en) 2003-08-13 2005-03-16 Japan Tobacco Inc Nitrogen-containing fused ring compound and use thereof as HIV integrase inhibitor
TW200528440A (en) 2003-10-31 2005-09-01 Fujisawa Pharmaceutical Co 2-cyanopyrrolidinecarboxamide compound
AU2005211349A1 (en) 2004-01-30 2005-08-18 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. N-benzyl-3,4-dihyroxypyridine-2-carboxamide and N-benzyl-2,3-dihydroxypyridine-4-carboxamide compounds useful as HIV integrase inhibitors
BRPI0507628A (en) 2004-02-11 2007-07-03 Smithkline Beecham Corp pharmaceutically acceptable compound or salt thereof, method of treating a viral infection in a human, use of a compound pharmaceutical composition, and process for preparing a compound
CN101014574A (en) 2004-03-09 2007-08-08 默克公司 HIV integrase inhibitors
WO2005110399A2 (en) 2004-04-29 2005-11-24 The Regents Of The University Of California Zinc-binding groups for metalloprotein inhibitors
EP1755586A2 (en) 2004-04-29 2007-02-28 The Regents of the University of California Hydroxypyridinone, hydroxypyridinethione, pyrone, and thiopyrone metalloprotein inhibitors
WO2005110414A2 (en) 2004-05-07 2005-11-24 Merck & Co., Inc. Hiv integrase inhibitors
US7273859B2 (en) 2004-05-12 2007-09-25 Bristol-Myers Squibb Company HIV integrase inhibitors: cyclic pyrimidinone compounds
CA2563601A1 (en) * 2004-05-17 2005-11-24 Tibotec Pharmaceuticals Ltd. Combinations of substituted 1-phenyl-1,5-dihydro-pyrido- [3,2-b] indol-2-ones and other hiv inhibitors
US7531554B2 (en) 2004-05-20 2009-05-12 Japan Tobacco Inc. 4-oxoquinoline compound and use thereof as HIV integrase inhibitor
MY134672A (en) 2004-05-20 2007-12-31 Japan Tobacco Inc Stable crystal of 4-oxoquinoline compound
EP2229945A1 (en) 2004-05-21 2010-09-22 Japan Tobacco, Inc. Combinations comprising a 4-isoquinolone derivative and anti-HIV agents
EP1790638B1 (en) 2004-09-15 2013-04-03 Shionogi Co., Ltd. Carbamoylpyridone derivative having hiv integrase inhibitory activity
JP2006118669A (en) 2004-10-25 2006-05-11 Sanoh Industrial Co Ltd Resin tube
CA2634499A1 (en) 2004-12-23 2006-06-29 Virochem Pharma Inc. Hydroxydihydropyridopy razine-1,8-diones and methods for inhibiting hiv integrase
EP1852434B1 (en) 2005-02-21 2011-07-13 Shionogi Co., Ltd. Bicyclic carbamoylpyridone derivative having hiv integrase inhibiting activity
EP1888581A2 (en) 2005-05-16 2008-02-20 Gilead Sciences, Inc. Hiv-integrase inhibitor compounds
TWI471145B (en) 2005-06-13 2015-02-01 Bristol Myers Squibb & Gilead Sciences Llc Unitary pharmaceutical dosage form
CA2616314A1 (en) 2005-07-27 2007-02-01 Gilead Sciences, Inc. Antiviral phosphonate conjugates for inhibition of hiv
CA2626956A1 (en) 2005-10-27 2007-05-03 Shionogi & Co., Ltd. Polycyclic carbamoylpyridone derivative having hiv integrase inhibitory activity
EP2308490A1 (en) 2005-12-30 2011-04-13 Gilead Sciences, Inc. Methods for improving the pharmacokinetics of hiv integrase inhibitors
US7601844B2 (en) 2006-01-27 2009-10-13 Bristol-Myers Squibb Company Piperidinyl derivatives as modulators of chemokine receptor activity
JP2009525261A (en) 2006-02-01 2009-07-09 日本たばこ産業株式会社 6- (3-Chloro-2-fluorobenzyl) -1-[(2S) -1-hydroxy-3-methylbutan-2-yl] -7-methoxy-4-oxo for the treatment of retroviral infections Use of -1,4-dihydroquinoline-3-carboxylic acid or a salt thereof
PL1992607T3 (en) 2006-03-06 2015-05-29 Japan Tobacco Inc Method for producing 4-oxoquinoline compound
CN101437801B (en) 2006-03-06 2013-02-06 日本烟草产业株式会社 Process for production of 4-oxoquinoline compound
US7893055B2 (en) 2006-06-28 2011-02-22 Bristol-Myers Squibb Company HIV integrase inhibitors
WO2008010953A2 (en) 2006-07-19 2008-01-24 University Of Georgia Research Foundation, Inc. Pyridinone diketo acids: inhibitors of hiv replication in combination therapy
MX2009002689A (en) 2006-09-12 2009-03-26 Gilead Sciences Inc Process and intermediates for preparing integrase inhibitors.
EP2084160A1 (en) 2006-10-18 2009-08-05 Merck & Co., Inc. Hiv integrase inhibitors
ES2603617T3 (en) 2007-02-23 2017-02-28 Gilead Sciences, Inc. Modulators of pharmacokinetic properties of therapeutics
US20080280945A1 (en) 2007-05-09 2008-11-13 Sachin Lohani Crystalline forms of an HIV integrase inhibitor
WO2009006203A1 (en) 2007-06-29 2009-01-08 Gilead Sciences, Inc. Therapeutic compositions and the use thereof
CN103480000A (en) 2007-06-29 2014-01-01 吉里德科学公司 Therapeutic compositions and the use thereof
US20090012103A1 (en) 2007-07-05 2009-01-08 Matthew Abelman Substituted heterocyclic compounds
WO2009018350A1 (en) 2007-07-31 2009-02-05 Limerick Biopharma, Inc. Pyrone analog compositions and methods
AR068403A1 (en) 2007-09-11 2009-11-18 Gilead Sciences Inc PROCESS AND INTERMEDIARIES FOR THE PREPARATION OF INTEGRASA INHIBITORS
EA201200631A1 (en) 2007-11-16 2012-11-30 Джилид Сайенсиз, Инк. Inhibitors of Human Immunodeficiency Virus Replication
GB0803019D0 (en) 2008-02-19 2008-03-26 Btg Int Ltd Fluorinated compounds
US8129398B2 (en) 2008-03-19 2012-03-06 Bristol-Myers Squibb Company HIV integrase inhibitors
US20100272811A1 (en) 2008-07-23 2010-10-28 Alkermes,Inc. Complex of trospium and pharmaceutical compositions thereof
EP2320909B8 (en) 2008-07-25 2016-03-30 VIIV Healthcare Company Chemical compounds
WO2010011815A1 (en) 2008-07-25 2010-01-28 Smithkline Beecham Corporation Chemical compounds
WO2010011819A1 (en) 2008-07-25 2010-01-28 Smithkline Beecham Corporation Chemical compounds
PT2320908E (en) 2008-07-25 2014-03-06 Shionogi & Co Dolutegravir prodrugs
WO2010011816A1 (en) * 2008-07-25 2010-01-28 Smithkline Beecham Corporation Chemical compounds
WO2010011818A1 (en) * 2008-07-25 2010-01-28 Smithkline Beecham Corporation Chemical compounds
ES2964383T3 (en) 2008-12-11 2024-04-05 Viiv Healthcare Co Processes and intermediates for carbamoylpyridone HIV integrase inhibitors
MX2011006241A (en) 2008-12-11 2011-06-28 Shionogi & Co Synthesis of carbamoylpyridone hiv integrase inhibitors and intermediates.
EP2367821B1 (en) 2008-12-17 2015-09-16 Merck Patent GmbH C-ring modified tricyclic benzonaphthiridinone protein kinase inhibitors and use thereof
AP3250A (en) 2009-02-06 2015-05-31 Gilead Sciences Inc Tablets for combination therapy
US8835461B2 (en) 2009-03-26 2014-09-16 Shionogi & Co., Ltd. Substituted 3-hydroxy-4-pyridone derivative
PT2444400T (en) 2009-06-15 2018-06-06 Shionogi & Co Substituted polycyclic carbamoylpyridone derivative
KR101280198B1 (en) 2009-09-02 2013-06-28 이화여자대학교 산학협력단 pyrazole derivatives, preparation thereof and composition comprising the same for prevention and treatment of osteoporosis
ES2688925T3 (en) 2010-01-27 2018-11-07 Viiv Healthcare Company Antiviral treatment
WO2011105590A1 (en) 2010-02-26 2011-09-01 日本たばこ産業株式会社 1,3,4,8-tetrahydro-2h-pyrido[1,2-a]pyrazine derivative and use of same as hiv integrase inhibitor
TWI582097B (en) 2010-03-23 2017-05-11 Viiv醫療保健公司 Process for preparing carbamoylpyridone derivatives and intermediates
US20130165489A1 (en) 2010-05-03 2013-06-27 The Trustees Of The University Of Pennsylvania Small Molecule Modulators of HIV-1 Capsid Stability and Methods Thereof
US9102614B2 (en) 2010-07-02 2015-08-11 Gilead Sciences, Inc. Naphth-2-ylacetic acid derivatives to treat AIDS
JP5806735B2 (en) 2010-07-02 2015-11-10 ギリアード サイエンシーズ, インコーポレイテッド 2-Quinolinyl-acetic acid derivatives as HIV antiviral compounds
EP2595986A2 (en) 2010-07-14 2013-05-29 Addex Pharma SA Novel 2-amino-4-pyrazolyl-thiazole derivatives and their use as allosteric modulators of metabotropic glutamate receptors
CN106083891B (en) 2010-08-05 2018-03-23 盐野义制药株式会社 The manufacture method of compound with hiv integrase inhibitory activity
SI2620436T1 (en) 2010-09-24 2018-08-31 Shionogi & Co., Ltd. Substituted polycyclic carbamoyl pyridone derivative prodrug
KR101923103B1 (en) 2010-11-19 2018-11-28 길리애드 사이언시즈, 인코포레이티드 Therapeutic compositions comprising rilpivirine hcl and tenofovir disoproxil fumarate
WO2012106534A2 (en) 2011-02-02 2012-08-09 The Regents Of The University Of California Hiv integrase inhibitors
AP2015008931A0 (en) 2011-04-21 2015-12-31 Gilead Sciences Inc Benzothiazole compounds and their pharmaceutical use
US20140213553A1 (en) 2011-05-03 2014-07-31 Concert Pharmaceuticals Inc. Carbamoylpyridone derivatives
US9328075B2 (en) 2011-05-05 2016-05-03 St. Jude Children's Research Hospital Pyrimidinone compounds and methods for treating influenza
US9121496B2 (en) 2011-06-29 2015-09-01 Arvinmeritor Technology, Llc Drive axle system and a method of control
US9540343B2 (en) 2011-07-06 2017-01-10 Gilead Sciences, Inc. Compounds for the treatment of HIV
RS56667B1 (en) 2011-07-07 2018-03-30 Janssen Sciences Ireland Uc Darunavir combination formulations
CN102863512B (en) 2011-07-07 2016-04-20 上海泓博智源医药技术有限公司 Antiviral compound
US9206197B2 (en) 2011-09-14 2015-12-08 Mapi Pharma Ltd. Amorphous form of dolutegravir
US9200009B2 (en) 2011-10-12 2015-12-01 Shionogi & Co., Ltd. Polycyclic pyridone derivative having integrase inhibitory activity
WO2013087581A1 (en) 2011-12-12 2013-06-20 Bayer Intellectual Property Gmbh Amino-substituted imidazopyridazines
US9284323B2 (en) 2012-01-04 2016-03-15 Gilead Sciences, Inc. Naphthalene acetic acid derivatives against HIV infection
WO2014008636A1 (en) 2012-07-11 2014-01-16 Merck Sharp & Dohme Corp. Macrocyclic compounds as hiv integrase inhibitors
WO2014014933A1 (en) 2012-07-20 2014-01-23 Merck Sharp & Dohme Corp. Hiv treatment with amido-substituted pyrimidinone derivatives
EP2877469A4 (en) 2012-07-25 2016-04-06 Merck Sharp & Dohme Substituted naphthyridinedione derivatives as hiv integrase inhibitors
US8877931B2 (en) 2012-08-03 2014-11-04 Gilead Sciences, Inc. Process and intermediates for preparing integrase inhibitors
WO2014074675A1 (en) 2012-11-08 2014-05-15 Bristol-Myers Squibb Company Heteroaryl substituted pyridyl compounds useful as kinase modulators
JP2016503030A (en) 2012-12-14 2016-02-01 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニーGlaxoSmithKline LLC Pharmaceutical composition
WO2014099586A1 (en) 2012-12-17 2014-06-26 Merck Sharp & Dohme Corp. 4-pyridinonetriazine derivatives as hiv integrase inhibitors
US20140221355A1 (en) 2012-12-21 2014-08-07 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
KR102527797B1 (en) 2012-12-21 2023-05-03 길리애드 사이언시즈, 인코포레이티드 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
EP2934482A4 (en) 2012-12-21 2016-07-20 Merck Sharp & Dohme Gastro-retentive formulations
US20140221380A1 (en) 2012-12-27 2014-08-07 Japan Tobacco Inc. SUBSTITUTED SPIROPYRIDO[1,2-a]PYRAZINE DERIVATIVE AND PHARMACEUTICAL USE OF SAME AS HIV INTEGRASE INHIBITOR
US9493479B2 (en) 2013-04-16 2016-11-15 Merck Sharp & Dohme Corp. Substituted pyrido[1,2-a]pyrazines as HIV integrase inhibitors
EP3008044B1 (en) 2013-06-13 2018-11-21 Merck Sharp & Dohme Corp. Fused tricyclic heterocyclic compounds as hiv integrase inhibitors
CA2916993C (en) * 2013-07-12 2019-01-15 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
NO2865735T3 (en) 2013-07-12 2018-07-21
WO2015039348A1 (en) 2013-09-23 2015-03-26 Merck Sharp & Dohme Corp. Tetracyclic heterocycle compounds useful as hiv integrase inhibitors
EA030695B1 (en) 2013-09-27 2018-09-28 Мерк Шарп И Доум Корп. Substituted quinolizine derivatives useful as hiv integrase inhibitors
WO2015089847A1 (en) 2013-12-20 2015-06-25 Merck Sharp & Dohme Corp. Spirocyclic heterocycle compounds useful as hiv integrase inhibitors
EP3096763B1 (en) 2014-01-21 2019-12-25 Laurus Labs Limited Novel process for the preparation of dolutegravir and pharmaceutically acceptable salts thereof
WO2015138933A1 (en) 2014-03-13 2015-09-17 Assia Chemical Industries Ltd. Solid state forms of dolutegravir sodium
TW201613936A (en) 2014-06-20 2016-04-16 Gilead Sciences Inc Crystalline forms of(2R,5S,13aR)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazepine-10-carboxamide
NO2717902T3 (en) 2014-06-20 2018-06-23
TWI744723B (en) 2014-06-20 2021-11-01 美商基利科學股份有限公司 Synthesis of polycyclic-carbamoylpyridone compounds
TWI733652B (en) * 2014-07-11 2021-07-21 美商基利科學股份有限公司 Modulators of toll-like receptors for the treatment of hiv
TWI695003B (en) 2014-12-23 2020-06-01 美商基利科學股份有限公司 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
AU2016244035B2 (en) 2015-04-02 2018-11-01 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
SI4070787T1 (en) 2015-06-30 2023-07-31 Gilead Sciences, Inc. Pharmaceutical formulations
CA2921336A1 (en) 2015-06-30 2016-12-30 Gilead Sciences, Inc. Pharmaceutical formulations
TWI737647B (en) 2015-11-09 2021-09-01 美商基利科學股份有限公司 Therapeutic compositions for treatment of human immunodeficiency virus

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006116764A1 (en) * 2005-04-28 2006-11-02 Smithkline Beecham Corporation Polycyclic carbamoylpyridone derivative having hiv integrase inhibitory activity
EP2412709A1 (en) * 2009-03-26 2012-02-01 Shionogi & Co., Ltd. Process for producing pyrone and pyridone derivatives

Cited By (302)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11034668B2 (en) 2011-07-06 2021-06-15 Gilead Sciences, Inc. Compounds for the treatment of HIV
US10428077B2 (en) 2012-04-26 2019-10-01 Bristol-Myers Squibb Company Imidazothiadiazole and imidazopyrazine derivatives as protease activated receptor4 (PAR4) inhibitors for treating platelet aggregation
US9688695B2 (en) 2012-04-26 2017-06-27 Bristol-Myers Squibb Company Imidazothiadiazole and imidazopyrazine derivatives as protease activated receptor 4 (PAR4) inhibitors for treating platelet aggregation
US10822343B2 (en) 2012-04-26 2020-11-03 Bristol-Myers Squibb Company Imidazothiadiazole and imidazopyrazine derivatives as protease activated receptor4 (PAR4) inhibitors for treating platelet aggregation
US10047103B2 (en) 2012-04-26 2018-08-14 Bristol-Myers Squibb Company Imidazothiadiazole and imidazopyrazine derivatives as protease activated receptor 4 (PAR4) inhibitors for treating platelet aggregation
US10821100B2 (en) 2012-06-15 2020-11-03 Ichnos Sciences SA Triazolone compounds as mPGES-1 inhibitors
US9949955B2 (en) 2012-06-15 2018-04-24 Glenmark Pharmaceuticals S.A. Triazolone compounds as mPGES-1 inhibitors
US10391083B2 (en) 2012-06-15 2019-08-27 Glenmark Pharmaceuticals S.A Triazolone compounds as MPGES-1 inhibitors
US10689399B2 (en) 2012-12-21 2020-06-23 Gilead Sciences, Inc. Substituted 3,4,5,6,8,10,14,14a-octahydro-2h-2,6-methanopyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazocines and methods for treating viral infections
US9732092B2 (en) 2012-12-21 2017-08-15 Gilead Sciences, Inc. Substituted 2,3,4,5,7,9,13,13a-octahydropyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]OXAZEPINES and methods for treating viral infections
US10246433B2 (en) 2012-12-21 2019-04-02 Pfizer Inc. Aryl and heteroaryl fused lactams
US10035809B2 (en) 2012-12-21 2018-07-31 Gilead Sciences, Inc. Substituted 2,3,4,5,7,9,13,13a-octahydro-1,5-methanopyrido[1′,2′:4,5]pyrazino[1,2-a][1,3]diazepines and methods for treating viral infections
US11548901B2 (en) 2012-12-21 2023-01-10 Gilead Sciences, Inc. Substituted 1,4-methanopyrido[1′,2′:4,5]pyrazino[1,2-a]pyrimidines for treating viral infections
US9663528B2 (en) 2012-12-21 2017-05-30 Gilead Sciences, Inc. Substituted 1,2,3,4,6,8,12,12a-octahydro-1,4-methanodipyrido[1,2-a:1',2'-d]pyrazines and methods for treating viral infections
US9216996B2 (en) 2012-12-21 2015-12-22 Gilead Sciences, Inc. Substituted 2,3,4,5,7,9,13,13a-octahydropyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazepines and methods for treating viral infections
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9849120B2 (en) 2013-01-15 2017-12-26 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10265307B2 (en) 2013-01-15 2019-04-23 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10828290B2 (en) 2013-01-15 2020-11-10 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as pim kinase inhibitors
US11229631B2 (en) 2013-01-15 2022-01-25 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10517858B2 (en) 2013-01-15 2019-12-31 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as PIM kinase inhibitors
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10456395B2 (en) 2013-07-12 2019-10-29 Gilead Sciences, Inc. Substituted dipyrido[1,2-a:1′,2′-d]pyrazines for treating viral infections
AU2018236731B2 (en) * 2013-07-12 2020-02-27 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their use for the treatment of HIV infections
WO2015006733A1 (en) * 2013-07-12 2015-01-15 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their use for the treatment of hiv infections
US11883397B2 (en) 2013-07-12 2024-01-30 Gilead Sciences, Inc. Substituted pyrido[1,2-a]pyrrolo[1,2-d]pyrazines for treating viral infections
EP3252053A1 (en) * 2013-07-12 2017-12-06 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their use for the treatment of hiv infections
AU2014286993B2 (en) * 2013-07-12 2018-10-25 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their use for the treatment of HIV infections
US10668064B2 (en) 2013-07-12 2020-06-02 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
JP2016525102A (en) * 2013-07-12 2016-08-22 ギリアード サイエンシス インコーポレーテッド Polycyclic carbamoylpyridone compounds and their use for treating HIV infection
JP2016527217A (en) * 2013-07-12 2016-09-08 ギリアード サイエンシス インコーポレーテッド Polycyclic carbamoylpyridone compounds and their use for treating HIV infection
EP3252058A1 (en) * 2013-07-12 2017-12-06 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their use for the treatment of hiv infections
US9458159B2 (en) 2013-07-12 2016-10-04 Gilead Sciences, Inc. Substituted pyrido[1′,2′:4,5]pyrazino[1,2-a]azepines for treating viral infections
US9421214B2 (en) 2013-07-12 2016-08-23 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
US9700554B2 (en) 2013-07-12 2017-07-11 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
WO2015006731A1 (en) * 2013-07-12 2015-01-15 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their use for the treatment of hiv infections
US11213523B2 (en) 2013-07-12 2022-01-04 Gilead Sciences, Inc. Substituted pyrido[1,2-a]pyrrolo[1,2-d]pyrazines for treating viral infections
AU2018236701B2 (en) * 2013-07-12 2020-04-30 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their use for the treatment of HIV infections
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
WO2015177537A1 (en) * 2014-05-20 2015-11-26 Cipla Limited Process for preparing polycyclic carbamoyl pyridone derivatives and intermediates thereof
US10570121B2 (en) 2014-06-17 2020-02-25 Pfizer Inc. Substituted dihydroisoquinolinone compounds
US10098886B2 (en) 2014-06-20 2018-10-16 Gilead Sciences, Inc. Crystalline forms of (2R,5S,13AR)-8-hydroxy-7,9-dioxo-N-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13A- octahydro-2,5-methanopyrido[1′,2′:4,5]pyrazino[2,1-B] [1,3] oxazepine-10-carboxamide
EA034169B1 (en) * 2014-06-20 2020-01-14 Джилид Сайэнс, Инк. Synthesis of polycyclic carbamoylpyridone compounds
KR20190049937A (en) * 2014-06-20 2019-05-09 길리애드 사이언시즈, 인코포레이티드 Synthesis of polycyclic-carbamoylpyridone compounds
KR101899803B1 (en) 2014-06-20 2018-09-20 길리애드 사이언시즈, 인코포레이티드 Sodium (2r,5s,13ar)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1',2':4.5]pyrazino[2,1-b]oxazepin-8-olate
EP4309736A2 (en) 2014-06-20 2024-01-24 Gilead Sciences, Inc. Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5]pyrazino [2,1-b][1,3]oxazepine-10-carboxamide
CN106459085A (en) * 2014-06-20 2017-02-22 吉利德科学公司 Sodium (2r, 5s, 13ar) -7, 9-dioxo-10- ( (2,4,6-trifluorobenzyl) carbamoyl) -2, 3, 4, 5, 7, 9, 13, 13a-octahydro-2, 5-methanopyrido [1',2' : 4.5] pyrazino [2, 1-b] oxazepin-8-olate
EP3783001A1 (en) * 2014-06-20 2021-02-24 Gilead Sciences, Inc. Synthesis of polycyclic-carbamoylpyridone compounds
EP4309736A3 (en) * 2014-06-20 2024-05-08 Gilead Sciences, Inc. Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5]pyrazino [2,1-b][1,3]oxazepine-10-carboxamide
JP2017521398A (en) * 2014-06-20 2017-08-03 ギリアード サイエンシス インコーポレーテッド Synthesis of polycyclic carbamoylpyridone compounds.
EP3157932B1 (en) 2014-06-20 2018-01-10 Gilead Sciences, Inc. Sodium (2r, 5s, 13ar) -7, 9-dioxo-10- ( (2,4,6-trifluorobenzyl) carbamoyl) -2, 3, 4, 5, 7, 9, 13, 13a-octahydro-2, 5-methanopyrido [1',2' : 4.5]pyrazino [2, 1-b]oxazepin-8-olate
WO2015195656A3 (en) * 2014-06-20 2016-03-24 Gilead Sciences ,Inc. Synthesis of polycyclic-carbamoylpyridone compounds
US11202780B2 (en) 2014-06-20 2021-12-21 Gilead Sciences, Inc. Crystalline forms of (2R,5S,13aR)-8-hydroxy-7,9-dioxo-N-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazepine-10-carboxamide
US9708342B2 (en) 2014-06-20 2017-07-18 Gilead Sciences, Inc. Sodium (2R,5S,13aR)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1′,2′:4,5]pyrazino[2, 1-b][1, 3]oxazepin-8-olate
MD20170006A2 (en) * 2014-06-20 2017-06-30 Gilead Sciences, Inc. Sodium (2R,5S,13aR)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazepin-8-olate
AU2018217236B2 (en) * 2014-06-20 2020-03-05 Gilead Sciences, Inc. Synthesis of polycyclic-carbamoylpyridone compounds
KR102159892B1 (en) 2014-06-20 2020-09-25 길리애드 사이언시즈, 인코포레이티드 Synthesis of polycyclic-carbamoylpyridone compounds
US9682084B2 (en) 2014-06-20 2017-06-20 Gilead Sciences, Inc. Crystalline forms of (2R,5S,13AR)-8-hydroxy-7,9,-dioxo-N-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazepine-10-carboxamide
AU2015277312B2 (en) * 2014-06-20 2018-08-30 Gilead Sciences, Inc. Synthesis of polycyclic-carbamoylpyridone compounds
CN114394983A (en) * 2014-06-20 2022-04-26 吉利德科学公司 Synthesis of polycyclic carbamoylpyridone compounds
AU2018203175B2 (en) * 2014-06-20 2020-01-02 Gilead Sciences, Inc. Sodium (2r,5s,13ar)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazepin-8-olate
US10385067B2 (en) 2014-06-20 2019-08-20 Gilead Sciences, Inc. Sodium (2R,5S,13aR)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazepin-8-olate
WO2015196137A1 (en) * 2014-06-20 2015-12-23 Gilead Sciences, Inc. Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5] pyrazino [2,1-b] [1,3] oxazepine-10-carboxamide
US10519168B2 (en) 2014-06-20 2019-12-31 Gilead Sciences, Inc. Synthesis of polycyclic-carbamoylpyridone compounds
EP3321270A1 (en) 2014-06-20 2018-05-16 Gilead Sciences, Inc. Sodium (2r, 5s, 13ar) -7, 9-dioxo-10- ( (2,4,6-trifluorobenzyl) carbamoyl) -2, 3, 4, 5, 7, 9, 13, 13a-octahydro-2, 5-methanopyrido [1',2' : 4.5] pyrazino [2, 1-b] oxazepin-8-olate
CN110317212B (en) * 2014-06-20 2022-10-21 吉利德科学公司 Synthesis of polycyclic carbamoylpyridone compounds
AU2015276860B2 (en) * 2014-06-20 2018-05-31 Gilead Sciences, Inc. Sodium (2r, 5s, 13ar) -7, 9-dioxo-10- ( (2,4,6-trifluorobenzyl) carbamoyl) -2, 3, 4, 5, 7, 9, 13, 13a-octahydro-2, 5-methanopyrido [1',2' : 4.5] pyrazino [2, 1-b][1,3]oxazepin-8-olate
EP3527573A1 (en) * 2014-06-20 2019-08-21 Gilead Sciences, Inc. Synthesis of polycyclic-carbamoylpyridone compounds
EP3564244A1 (en) 2014-06-20 2019-11-06 Gilead Sciences, Inc. Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5]pyrazino [2,1-b][1,3]oxazepine-10-carboxamide
WO2015196116A1 (en) * 2014-06-20 2015-12-23 Gilead Sciences, Inc. Sodium (2r, 5s, 13ar) -7, 9-dioxo-10- ( (2,4,6-trifluorobenzyl) carbamoyl) -2, 3, 4, 5, 7, 9, 13, 13a-octahydro-2, 5-methanopyrido [1',2' : 4.5] pyrazino [2, 1-b] oxazepin-8-olate
EA030967B1 (en) * 2014-06-20 2018-10-31 Джилид Сайэнс, Инк. SODIUM (2R,5S,13aR)-7,9-DIOXO-10-((2,4,6-TRIFLUOROBENZYL)CARBAMOYL)-2,3,4,5,7,9,13,13a-OCTAHYDRO-2,5-METHANOPYRIDO[1',2':4,5]PYRAZINO[2,1-b][1,3]OXAZEPIN-8-OLATE
US10975096B2 (en) 2014-06-20 2021-04-13 Gilead Sciences, Inc. Synthesis of polycyclic-carbamoylpyridone compounds
CN110317212A (en) * 2014-06-20 2019-10-11 吉利德科学公司 The synthesis of polycyclic carbamoylpyridone compound
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
JPWO2016027879A1 (en) * 2014-08-22 2017-06-01 塩野義製薬株式会社 Polycyclic pyridone derivatives having integrase inhibitory activity
US10011613B2 (en) 2014-08-22 2018-07-03 Shionogi & Co., Ltd. Polycyclic pyridone derivative having integrase inhibitory activity
US10202404B2 (en) 2014-08-22 2019-02-12 Shionogi & Co., Ltd. Polycyclic pyridone derivative having integrase inhibitory activity
JP2021042237A (en) * 2014-08-22 2021-03-18 塩野義製薬株式会社 Polycyclic pyridone derivative having integrase inhibitory activity
EP3757105A2 (en) 2014-08-22 2020-12-30 Shionogi & Co., Ltd Polycyclic pyridone derivative having integrase inhibitory activity
WO2016027879A1 (en) * 2014-08-22 2016-02-25 塩野義製薬株式会社 Polycyclic pyridone derivative having integrase-inhibiting activity
WO2016036759A1 (en) 2014-09-04 2016-03-10 Gilead Sciences, Inc. Methods of treating or preventing hiv in patients using a combination of tenofovir alafenamide and dolutegravir
US9795602B2 (en) 2014-12-23 2017-10-24 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
US9522912B2 (en) 2014-12-23 2016-12-20 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
KR102020004B1 (en) 2014-12-23 2019-09-06 길리애드 사이언시즈, 인코포레이티드 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
JP2018172437A (en) * 2014-12-23 2018-11-08 ギリアード サイエンシーズ, インコーポレイテッド Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
EP3388431A1 (en) 2014-12-23 2018-10-17 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
KR20190052172A (en) * 2014-12-23 2019-05-15 길리애드 사이언시즈, 인코포레이티드 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
WO2016106237A1 (en) 2014-12-23 2016-06-30 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
KR20190104458A (en) * 2014-12-23 2019-09-09 길리애드 사이언시즈, 인코포레이티드 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
JP2017538713A (en) * 2014-12-23 2017-12-28 ギリアード サイエンシーズ, インコーポレイテッド Polycyclic carbamoylpyridone compounds and their pharmaceutical use
CN111454273A (en) * 2014-12-23 2020-07-28 吉利德科学公司 Polycyclic carbonyl pyridinone compounds and pharmaceutical use thereof
KR20170097168A (en) * 2014-12-23 2017-08-25 길리애드 사이언시즈, 인코포레이티드 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
KR102240584B1 (en) 2014-12-23 2021-04-15 길리애드 사이언시즈, 인코포레이티드 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
CN107108643A (en) * 2014-12-23 2017-08-29 吉利德科学公司 Polycyclic carbonic acyl radical pyridinone compounds and its medicinal usage
US10646486B2 (en) 2014-12-23 2020-05-12 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
KR101978167B1 (en) 2014-12-23 2019-05-14 길리애드 사이언시즈, 인코포레이티드 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
US10206926B2 (en) 2014-12-24 2019-02-19 Gilead Sciences, Inc. Quinazoline compounds
US10548898B2 (en) 2014-12-24 2020-02-04 Gilead Sciences Inc. Quinazoline compounds
EP3521282A1 (en) 2014-12-24 2019-08-07 Gilead Sciences, Inc. Quinazoline derivatives used to treat hiv
WO2016105564A1 (en) 2014-12-24 2016-06-30 Gilead Sciences, Inc. Quinazoline derivatives used to treat hiv
WO2016105534A1 (en) 2014-12-24 2016-06-30 Gilead Sciences, Inc. Isoquinoline compounds for the treatment of hiv
US11304948B2 (en) 2014-12-24 2022-04-19 Gilead Sciences, Inc. Quinazoline compounds
WO2016105532A1 (en) 2014-12-24 2016-06-30 Gilead Sciences, Inc. Fused pyrimidine compounds for the treatment of hiv
EP3960735A1 (en) 2014-12-24 2022-03-02 Gilead Sciences, Inc. Quinazoline derivatives used to treat hiv
US9730936B2 (en) 2014-12-24 2017-08-15 Gilead Sciences, Inc. Quinazoline compounds
US9701677B2 (en) 2014-12-24 2017-07-11 Gilead Sciences, Inc. Fused pyrimidine compounds
EP4302830A2 (en) 2014-12-24 2024-01-10 Gilead Sciences, Inc. Quinazoline derivatives used to treat hiv
US9624195B2 (en) 2014-12-24 2017-04-18 Gilead Sciences, Inc. Isoquinoline compounds
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2016141092A1 (en) 2015-03-04 2016-09-09 Gilead Sciences, Inc. Toll-like receptor modulating 4,6-diamino-pyrido[3,2-d]pyrimidine compounds
EP3722297A1 (en) 2015-03-04 2020-10-14 Gilead Sciences, Inc. Toll-like receptor modulating 4,6-diamino-pyrido[3,2-d]pyrimidine compounds
EP3321265A1 (en) 2015-03-04 2018-05-16 Gilead Sciences, Inc. 4,6-diamino-pyrido[3,2-d]pyrimidine compounds and their utilisation as modulators of toll-like receptors
EP3736274A1 (en) 2015-04-02 2020-11-11 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
JP2018188483A (en) * 2015-04-02 2018-11-29 ギリアード サイエンシーズ, インコーポレイテッド Polycyclic carbamoylpyridone compound and pharmaceutical use thereof
JP2018510168A (en) * 2015-04-02 2018-04-12 ギリアード サイエンシーズ, インコーポレイテッド Polycyclic carbamoylpyridone compounds and their pharmaceutical use
WO2016161382A1 (en) 2015-04-02 2016-10-06 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
RU2712275C9 (en) * 2015-04-28 2020-03-19 Сионоги Энд Ко., Лтд. Substituted polycyclic pyridone derivatives and prodrugs thereof
RU2712275C2 (en) * 2015-04-28 2020-01-28 Сионоги Энд Ко., Лтд. Substituted polycyclic pyridone derivatives and prodrugs thereof
US9802918B2 (en) 2015-05-29 2017-10-31 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
WO2017035230A1 (en) 2015-08-26 2017-03-02 Gilead Sciences, Inc. Deuterated toll-like receptor modulators
US11066387B2 (en) 2015-09-09 2021-07-20 Incyte Corporation Salts of a Pim kinase inhibitor
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
US11505540B2 (en) 2015-09-09 2022-11-22 Incyte Corporation Salts of a Pim kinase inhibitor
US10336728B2 (en) 2015-09-09 2019-07-02 Incyte Corporation Salts of a Pim kinase inhibitor
WO2017059120A1 (en) 2015-09-30 2017-04-06 Gilead Sciences, Inc. Compounds and combinations for the treatment of hiv
US11053215B2 (en) 2015-10-02 2021-07-06 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US10450296B2 (en) 2015-10-02 2019-10-22 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
CN108348473A (en) * 2015-11-09 2018-07-31 吉利德科学公司 Treat the therapeutic combination of human immunodeficiency virus
TWI737647B (en) * 2015-11-09 2021-09-01 美商基利科學股份有限公司 Therapeutic compositions for treatment of human immunodeficiency virus
KR102606625B1 (en) * 2015-11-09 2023-11-27 길리애드 사이언시즈, 인코포레이티드 Therapeutic Compositions for Treatment of Human Immunodeficiency Virus
US10548846B2 (en) 2015-11-09 2020-02-04 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
US11744802B2 (en) 2015-11-09 2023-09-05 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
AU2016354007C9 (en) * 2015-11-09 2020-10-01 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
EP3346995B1 (en) 2015-11-09 2019-08-28 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
KR20200106222A (en) * 2015-11-09 2020-09-11 길리애드 사이언시즈, 인코포레이티드 Therapeutic Compositions for Treatment of Human Immunodeficiency Virus
JP2018532811A (en) * 2015-11-09 2018-11-08 ギリアード サイエンシス インコーポレーテッド Therapeutic composition for treating human immunodeficiency virus
AU2016354007C1 (en) * 2015-11-09 2020-09-10 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
AU2020200995B9 (en) * 2015-11-09 2022-04-28 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
KR102153996B1 (en) * 2015-11-09 2020-09-09 길리애드 사이언시즈, 인코포레이티드 Therapeutic composition for the treatment of human immunodeficiency virus
EP3632415A1 (en) 2015-11-09 2020-04-08 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
CN108348473B (en) * 2015-11-09 2021-06-18 吉利德科学公司 Therapeutic compositions for the treatment of human immunodeficiency virus
KR20180067702A (en) * 2015-11-09 2018-06-20 길리애드 사이언시즈, 인코포레이티드 Therapeutic composition for the treatment of human immunodeficiency virus
WO2017083304A1 (en) 2015-11-09 2017-05-18 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
CN113546052A (en) * 2015-11-09 2021-10-26 吉利德科学公司 Therapeutic compositions for the treatment of human immunodeficiency virus
AU2016354007B2 (en) * 2015-11-09 2019-11-14 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
AU2020200995B2 (en) * 2015-11-09 2022-04-07 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
WO2017106346A2 (en) 2015-12-15 2017-06-22 Gilead Sciences, Inc. Human immunodeficiency virus neutralizing antibodies
EP3992206A1 (en) 2015-12-15 2022-05-04 Gilead Sciences, Inc. Human immunodeficiency virus neutralizing antibodies
EP3597646A1 (en) 2016-08-19 2020-01-22 Gilead Sciences, Inc. Therapeutic compounds useful for the prophylactic or therapeutic treatment of an hiv virus infection
US11993583B2 (en) 2016-08-19 2024-05-28 Gilead Sciences, Inc. Therapeutic compounds
US10654827B2 (en) 2016-08-19 2020-05-19 Gilead Sciences, Inc. Therapeutic compounds
EP4265299A2 (en) 2016-08-19 2023-10-25 Gilead Sciences, Inc. Therapeutic compounds useful for the prophylactic or therapeutic treatment of an hiv virus infection
WO2018035359A1 (en) 2016-08-19 2018-02-22 Gilead Sciences, Inc. Therapeutic compounds useful for the prophylactic or therapeutic treatment of an hiv virus infection
WO2018042332A1 (en) 2016-08-31 2018-03-08 Glaxosmithkline Intellectual Property (No.2) Limited Combinations and uses and treatments thereof
WO2018042331A1 (en) 2016-08-31 2018-03-08 Glaxosmithkline Intellectual Property (No.2) Limited Combinations and uses and treatments thereof
WO2018045144A1 (en) 2016-09-02 2018-03-08 Gilead Sciences, Inc. Toll like receptor modulator compounds
WO2018045150A1 (en) 2016-09-02 2018-03-08 Gilead Sciences, Inc. 4,6-diamino-pyrido[3,2-d]pyrimidine derivaties as toll like receptor modulators
WO2018051250A1 (en) 2016-09-14 2018-03-22 Viiv Healthcare Company Combination comprising tenofovir alafenamide, bictegravir and 3tc
WO2018064071A1 (en) 2016-09-27 2018-04-05 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
WO2018064080A1 (en) 2016-09-28 2018-04-05 Gilead Sciences, Inc. Benzothiazol-6-yl acetic acid derivatives and their use for treating hiv infection
WO2018081292A1 (en) 2016-10-27 2018-05-03 Gilead Sciences, Inc. Crystalline forms of darunavir free base, hydrate, solvates and salts
US10829499B2 (en) 2016-12-02 2020-11-10 Merck Sharp & Dohme Corp. Tetracyclic heterocycle compounds useful as HIV integrase inhibitors
US10479801B2 (en) 2016-12-02 2019-11-19 Merck Sharp & Dohme Corp. Tetracyclic heterocycle compounds useful as HIV integrase inhibitors
WO2018144390A1 (en) 2017-01-31 2018-08-09 Gilead Sciences, Inc. Crystalline forms of tenofovir alafenamide
US10752636B2 (en) 2017-02-06 2020-08-25 Gilead Sciences, Inc. HIV inhibitor compounds
EP3909949A1 (en) 2017-02-06 2021-11-17 Gilead Sciences, Inc. Atazanavir (atv) analogues for treating hiv infections
WO2018145021A1 (en) 2017-02-06 2018-08-09 Gilead Sciences, Inc. Atazanavir (atv) analogues for treating hiv infections
US11078208B1 (en) 2017-02-06 2021-08-03 Gilead Sciences, Inc. HIV inhibitor compounds
WO2018229798A1 (en) 2017-06-13 2018-12-20 Cipla Limited Process for the preparation of bictegravir and intermediate thereof
WO2018237148A1 (en) 2017-06-21 2018-12-27 Gilead Sciences, Inc. Multispecific antibodies that target hiv gp120 and cd3
WO2019027920A1 (en) 2017-08-01 2019-02-07 Gilead Sciences, Inc. Crystalline forms of ethyl ((s)-((((2r,5r)-5-(6-amino-9h-purin-9-yl)-4-fluoro-2,5-dihydrofuran-2-yl)oxy)methyl)(phenoxy)phosphoryl)-l-alaninate (gs-9131) for treating viral infections
US10851125B2 (en) 2017-08-01 2020-12-01 Gilead Sciences, Inc. Crystalline forms of ethyl ((S)-((((2R,5R)-5-(6-amino-9H-purin-9-yl)-4-fluoro-2,5-dihydrofuran-2-yl)oxy)methyl)(phenoxy)phosphoryl(-L-alaninate
EP3960740A1 (en) 2017-08-01 2022-03-02 Gilead Sciences, Inc. Crystalline forms of ethyl ((s)-((((2r,5r)-5-(6-amino-9h-purin-9-yl)-4-fluoro-2,5-dihydrofuran-2-yl)oxy)methyl)(phenoxy)phosphoryl)-l-alaninate (gs-9131) vanillate for treating viral infections
EP3664896A1 (en) 2017-08-09 2020-06-17 VIIV Healthcare Company Combinations and uses and treatments thereof
EP3664895A1 (en) 2017-08-09 2020-06-17 VIIV Healthcare Company Combinations and uses and treatments thereof
EP3664896A4 (en) * 2017-08-09 2021-06-09 VIIV Healthcare Company Combinations and uses and treatments thereof
US11845739B2 (en) 2017-08-17 2023-12-19 Gilead Sciences, Inc. Solid forms of an HIV capsid inhibitor
EP4092020A1 (en) 2017-08-17 2022-11-23 Gilead Sciences, Inc. Solid forms of an hiv capsid inhibitor
US11267799B2 (en) 2017-08-17 2022-03-08 Gilead Sciences, Inc. Solid forms of an HIV capsid inhibitor
US10849892B2 (en) 2017-08-17 2020-12-01 Gilead Sciences, Inc. Choline salt forms of an HIV capsid inhibitor
WO2019035973A1 (en) 2017-08-17 2019-02-21 Gilead Sciences, Inc Choline salt forms of an hiv capsid inhibitor
US11266638B2 (en) 2017-08-17 2022-03-08 Gilead Sciences, Inc. Choline salt forms of an HIV capsid inhibitor
EP4382105A2 (en) 2017-08-17 2024-06-12 Gilead Sciences, Inc. Choline salt forms of an hiv capsid inhibitor
EP4046994A1 (en) 2017-08-17 2022-08-24 Gilead Sciences, Inc. Choline salt forms of an hiv capsid inhibitor
US11833143B2 (en) 2017-08-17 2023-12-05 Gilead Sciences, Inc. Choline salt forms of an HIV capsid inhibitor
WO2019035904A1 (en) 2017-08-17 2019-02-21 Gilead Sciences, Inc. Solid forms of an hiv capsid inhibitor
US11286262B2 (en) 2017-10-06 2022-03-29 Shionogi & Co., Ltd. Stereoselective process for preparing substituted polycyclic pyridone derivatives
EP3693373A4 (en) * 2017-10-06 2021-11-03 Shionogi & Co., Ltd. Method for stereoselectively producing substituted polycyclic pyridone derivative
WO2019075291A1 (en) 2017-10-13 2019-04-18 Gilead Sciences, Inc. 1-benzyl-2-imino-4-phenyl-5-oxoimidazolidine derivatives as hiv protease inhibitors
WO2019084020A1 (en) 2017-10-24 2019-05-02 Gilead Sciences, Inc. Methods of treating patients co-infected with a virus and tuberculosis
WO2019113462A1 (en) 2017-12-07 2019-06-13 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11331331B2 (en) 2017-12-07 2022-05-17 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11903959B2 (en) 2017-12-07 2024-02-20 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11278541B2 (en) 2017-12-08 2022-03-22 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US11203610B2 (en) 2017-12-20 2021-12-21 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
US10966999B2 (en) 2017-12-20 2021-04-06 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
US11253524B2 (en) 2018-01-19 2022-02-22 Gilead Sciences, Inc. Metabolites of bictegravir
WO2019154634A1 (en) 2018-02-09 2019-08-15 Sandoz Ag Crystalline form of bictegravir sodium
WO2019161017A1 (en) 2018-02-15 2019-08-22 Gilead Sciences, Inc. Pyridine derivatives and their use for treating hiv infection
US10836746B2 (en) 2018-02-15 2020-11-17 Gilead Sciences, Inc. Therapeutic compounds
US11267801B2 (en) 2018-02-15 2022-03-08 Gilead Sciences, Inc. Therapeutic compounds
US11753399B2 (en) 2018-02-15 2023-09-12 Gilead Sciences, Inc. Therapeutic compounds
WO2019159199A1 (en) 2018-02-16 2019-08-22 Cipla Limited Continues flow process for the preparation of active pharmaceutical ingredients - polycyclic carbamoyl pyridone derivatives and intermediates thereof
US10696657B2 (en) 2018-02-16 2020-06-30 Gilead Sciences, Inc. Methods and intermediates for preparing therapeutic compounds
US11760746B2 (en) 2018-02-16 2023-09-19 Gilead Sciences, Inc. Methods and intermediates for preparing therapeutic compounds
US11117886B2 (en) 2018-02-16 2021-09-14 Gilead Sciences, Inc. Methods and intermediates for preparing therapeutic compounds
WO2019193543A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
US11292812B2 (en) 2018-04-06 2022-04-05 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′-cyclic dinucleotides
WO2019193533A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'2'-cyclic dinucleotides
US11149052B2 (en) 2018-04-06 2021-10-19 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′-cyclic dinucleotides
WO2019193542A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides
WO2019211799A1 (en) 2018-05-03 2019-11-07 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide
EP4257137A2 (en) 2018-05-31 2023-10-11 Shionogi & Co., Ltd Polycyclic carbamoylpyridone derivatives for the treatment of hiv
US11453669B2 (en) 2018-05-31 2022-09-27 Shionogi & Co., Ltd. Polycyclic pyridone derivative
KR20210015922A (en) 2018-05-31 2021-02-10 시오노기 앤드 컴파니, 리미티드 Polycyclic carbamoylpyridone derivatives
US11649236B2 (en) 2018-05-31 2023-05-16 Shionogi & Co., Ltd. Polycyclic carbamoylpyridone derivative
WO2019230858A1 (en) 2018-05-31 2019-12-05 塩野義製薬株式会社 Polycyclic carbamoylpyridone derivative
WO2020004443A1 (en) 2018-06-27 2020-01-02 国立大学法人北海道大学 Arenavirus growth inhibitor comprising polycyclic carbamoyl pyridone derivative
US11623933B2 (en) 2018-06-28 2023-04-11 Honour Lab Limited Process for the preparation of sodium (2R,5S,13AR)-7,9-dioxo-10-((2,4,6-trifluorobenzyl) carbamoyl)-2,3,4,5,7,9,13,13A-octahydro-2,5-Methanopyrido[1 ′,2′:4,5]pyrazino[2,1 -b] [1,3] oxazepin-8-olate and its polymorphic form
WO2020003151A1 (en) * 2018-06-28 2020-01-02 Honour Lab Limited Process for the preparation of sodium (2r,5s,13ar)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1', 2':4,5]pyrazino[2,1-b] [1,3] oxazepin-8-olate and its polymorphic form
EP4257600A2 (en) 2018-07-03 2023-10-11 Gilead Sciences, Inc. Antibodies that target hiv gp120 and methods of use
WO2020010107A1 (en) 2018-07-03 2020-01-09 Gilead Sciences, Inc. Antibodies that target hiv gp120 and methods of use
WO2020012408A3 (en) * 2018-07-12 2020-03-05 Laurus Labs Limited A process for purification of protected polycyclic carbamoylpyridone derivatives
US11634431B2 (en) 2018-07-12 2023-04-25 Laurus Labs Limited Process for purification of protected polycyclic carbamoylpyridone derivatives
US11944611B2 (en) 2018-07-16 2024-04-02 Gilead Sciences, Inc. Capsid inhibitors for the treatment of HIV
WO2020018459A1 (en) 2018-07-16 2020-01-23 Gilead Sciences, Inc. Capsid inhibitors for the treatment of hiv
WO2020028272A1 (en) 2018-07-30 2020-02-06 Gilead Sciences, Inc. Anti-hiv compounds
US11052087B2 (en) 2018-07-30 2021-07-06 Gilead Sciences, Inc. Anti-HIV compounds
WO2020072656A1 (en) 2018-10-03 2020-04-09 Gilead Sciences, Inc. Imidozopyrimidine derivatives
EP4371987A1 (en) 2018-10-31 2024-05-22 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
WO2020092528A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity
WO2020092621A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
EP3653629A1 (en) 2018-11-16 2020-05-20 Sandoz AG Acid addition salts of an integrase strand transfer inhibitor
WO2020161744A1 (en) 2019-02-07 2020-08-13 Cipla Limited Novel polymorphs of integrase inhibitor
WO2020176510A1 (en) 2019-02-25 2020-09-03 Gilead Sciences, Inc. Protein kinase c agonists
WO2020176505A1 (en) 2019-02-25 2020-09-03 Gilead Sciences, Inc. Protein kinase c agonists
WO2020178768A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
WO2020178769A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides and prodrugs thereof
WO2020178770A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides and prodrugs thereof
US11766447B2 (en) 2019-03-07 2023-09-26 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
TWI745900B (en) * 2019-03-22 2021-11-11 美商基利科學股份有限公司 Bridged tricyclic carbamoylpyridone compounds and their pharmaceutical use
WO2020197991A1 (en) 2019-03-22 2020-10-01 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and their pharmaceutical use
US11084832B2 (en) 2019-03-22 2021-08-10 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and their pharmaceutical use
EP4122537A1 (en) 2019-03-22 2023-01-25 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and their pharmaceutical use
US11548902B1 (en) 2019-03-22 2023-01-10 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and their pharmaceutical use
WO2020214716A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. 2-imino-5-oxo-imidazolidine inhibitors of hiv protease
WO2020214663A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020214647A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of an hiv protease inhibitor
WO2020214652A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020236753A1 (en) 2019-05-21 2020-11-26 Gilead Sciences, Inc. Methods of identifying hiv patients sensitive to therapy with gp120 v3 glycan-directed antibodies
WO2020237025A1 (en) 2019-05-23 2020-11-26 Gilead Sciences, Inc. Substituted exo-methylene-oxindoles which are hpk1/map4k1 inhibitors
EP3978503A4 (en) * 2019-06-03 2022-12-07 Ivachtchenko, Alena Alexandrovna ANNELATED 9-HYDROXY-1,8-DIOXO-1,3,4,8-TETRAHYDRO-2H-PYRIDO[1,2-a]PYRAZINE-7-CARBOXAMIDES AS HIV INTEGRASE INHIBITORS
RU2717101C1 (en) * 2019-06-03 2020-03-18 Андрей Александрович Иващенко Anellated 9-hydroxy-1,8-dioxo-1,3,4,8-tetrahydro-2h-pyrido[1,2-a]pyrazine-7-carboxamides - integrase inhibitors, methods for preparing and using thereof
WO2020246910A1 (en) 2019-06-03 2020-12-10 Александл Васильевич ИВАЩЕНКО ANNELATED 9-HYDROXY-1,8-DIOXO-1,3,4,8-TETRAHYDRO-2Н-PYRIDO[1,2-α]PYRAZINE-7-CARBOXAMIDES AS HIV INTEGRASE INHIBITORS
WO2020255038A1 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company Combination of hepatitis b virus (hbv) vaccines and pyridopyrimidine derivatives
EP3993797A4 (en) * 2019-06-18 2023-10-25 Laurus Labs Limited Process and polymorphic forms of bictegravir and its pharmaceutically acceptable salts or co-crystals thereof
US11358711B2 (en) 2019-06-20 2022-06-14 Merck Sharp & Dohme Corp. Tetracyclic heterocycle compounds useful as HIV integrase inhibitors
WO2020263830A1 (en) 2019-06-25 2020-12-30 Gilead Sciences, Inc. Flt3l-fc fusion proteins and methods of use
US11248005B2 (en) 2019-07-08 2022-02-15 Lupin Limited Process for preparation of intermediates used for the synthesis of HIV integrase inhibitor
WO2021011544A1 (en) 2019-07-16 2021-01-21 Gilead Sciences, Inc. Hiv vaccines and methods of making and using
WO2021011891A1 (en) 2019-07-18 2021-01-21 Gilead Sciences, Inc. Long-acting formulations of tenofovir alafenamide
WO2021034804A1 (en) 2019-08-19 2021-02-25 Gilead Sciences, Inc. Pharmaceutical formulations of tenofovir alafenamide
US11807625B2 (en) 2019-11-26 2023-11-07 Gilead Sciences, Inc. Capsid inhibitors for the prevention of HIV
WO2021107066A1 (en) 2019-11-28 2021-06-03 塩野義製薬株式会社 Prophylactic and therapeutic pharmaceutical agent for hiv infectious diseases characterized by comprising combination of integrase inhibitor and anti-hiv agent
WO2021130638A1 (en) 2019-12-24 2021-07-01 Carna Biosciences, Inc. Diacylglycerol kinase modulating compounds
US11697652B2 (en) 2020-02-24 2023-07-11 Gilead Sciences, Inc. Tetracyclic compounds and uses thereof
WO2021173522A1 (en) 2020-02-24 2021-09-02 Gilead Sciences, Inc. Tetracyclic compounds for treating hiv infection
WO2021188959A1 (en) 2020-03-20 2021-09-23 Gilead Sciences, Inc. Prodrugs of 4'-c-substituted-2-halo-2'-deoxyadenosine nucleosides and methods of making and using the same
WO2021236944A1 (en) 2020-05-21 2021-11-25 Gilead Sciences, Inc. Pharmaceutical compositions comprising bictegravir
CN113698420A (en) * 2020-05-22 2021-11-26 上海迪赛诺生物医药有限公司 Novel crystal form of bicalutavir sodium and preparation method thereof
US11680064B2 (en) 2020-06-25 2023-06-20 Gilead Sciences, Inc. Capsid inhibitors for the treatment of HIV
WO2021262990A1 (en) 2020-06-25 2021-12-30 Gilead Sciences, Inc. Capsid inhibitors for the treatment of hiv
WO2022031894A1 (en) 2020-08-07 2022-02-10 Gilead Sciences, Inc. Prodrugs of phosphonamide nucleotide analogues and their pharmaceutical use
WO2022072520A1 (en) 2020-09-30 2022-04-07 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and uses thereof
WO2022087149A2 (en) 2020-10-22 2022-04-28 Gilead Sciences, Inc. Interleukin-2-fc fusion proteins and methods of use
CN114426540A (en) * 2020-10-29 2022-05-03 上海拓界生物医药科技有限公司 Pyrido [1,2-a ] pyrazine-1, 8-diketone prodrug derivatives, preparation method and application thereof
CN114426540B (en) * 2020-10-29 2024-04-26 上海拓界生物医药科技有限公司 Pyrido [1,2-a ] pyrazine-1, 8-dione prodrug derivatives, preparation method and application thereof
WO2022103758A1 (en) 2020-11-11 2022-05-19 Gilead Sciences, Inc. METHODS OF IDENTIFYING HIV PATIENTS SENSITIVE TO THERAPY WITH gp120 CD4 BINDING SITE-DIRECTED ANTIBODIES
EP4321217A2 (en) 2021-01-19 2024-02-14 Gilead Sciences, Inc. Substituted pyridotriazine compounds and uses thereof
WO2022159387A1 (en) 2021-01-19 2022-07-28 Gilead Sciences, Inc. Substituted pyridotriazine compounds and uses thereof
US11613546B2 (en) 2021-01-19 2023-03-28 Gilead Sciences, Inc. Substituted pyridotriazine compounds and uses thereof
US11897892B2 (en) 2021-01-19 2024-02-13 Gilead Sciences, Inc. Substituted pyridotriazine compounds and uses thereof
WO2022245671A1 (en) 2021-05-18 2022-11-24 Gilead Sciences, Inc. Methods of using flt3l-fc fusion proteins
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2023102523A1 (en) 2021-12-03 2023-06-08 Gilead Sciences, Inc. Therapeutic compounds for hiv virus infection
WO2023102239A1 (en) 2021-12-03 2023-06-08 Gilead Sciences, Inc. Therapeutic compounds for hiv virus infection
WO2023102529A1 (en) 2021-12-03 2023-06-08 Gilead Sciences, Inc. Therapeutic compounds for hiv virus infection
US11787825B2 (en) 2021-12-03 2023-10-17 Gilead Sciences, Inc. Therapeutic compounds for HIV virus infection
WO2023196875A1 (en) 2022-04-06 2023-10-12 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and uses thereof
EP4310087A1 (en) 2022-04-06 2024-01-24 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and uses thereof
WO2024006982A1 (en) 2022-07-01 2024-01-04 Gilead Sciences, Inc. Therapeutic compounds useful for the prophylactic or therapeutic treatment of an hiv virus infection
WO2024015741A1 (en) 2022-07-12 2024-01-18 Gilead Sciences, Inc. Hiv immunogenic polypeptides and vaccines and uses thereof
WO2024044477A1 (en) 2022-08-26 2024-02-29 Gilead Sciences, Inc. Dosing and scheduling regimen for broadly neutralizing antibodies
WO2024076915A1 (en) 2022-10-04 2024-04-11 Gilead Sciences, Inc. 4'-thionucleoside analogues and their pharmaceutical use
US12024528B2 (en) 2023-06-14 2024-07-02 Gilead Sciences, Inc. Bridged tricyclic carbamoylpyridone compounds and uses thereof

Also Published As

Publication number Publication date
LTPA2018511I1 (en) 2018-10-10
IL239316A0 (en) 2015-07-30
AU2018232957A1 (en) 2018-10-11
LUC00083I2 (en) 2018-10-15
ME02400B (en) 2016-09-20
MX2020003055A (en) 2020-07-27
EP4122935A1 (en) 2023-01-25
IL245780A0 (en) 2016-08-02
AU2021221427A1 (en) 2021-09-16
NZ718708A (en) 2018-11-30
LT3608325T (en) 2022-09-12
TW201918485A (en) 2019-05-16
CY2018022I1 (en) 2019-07-10
PH12015501445A1 (en) 2015-09-14
CN104995198A (en) 2015-10-21
ES2926068T3 (en) 2022-10-21
EA037633B1 (en) 2021-04-23
IL247311A0 (en) 2016-09-29
MD20200093A2 (en) 2021-05-31
US10689399B2 (en) 2020-06-23
CR20170279A (en) 2017-10-19
KR102246963B1 (en) 2021-04-30
JP6933690B2 (en) 2021-09-08
MX2015008009A (en) 2015-10-22
CY1117570T1 (en) 2017-04-26
CN107674086A (en) 2018-02-09
KR102040007B1 (en) 2019-11-05
TW202400591A (en) 2024-01-01
UA114351C2 (en) 2017-05-25
MD4841B1 (en) 2023-01-31
CN116640140A (en) 2023-08-25
JP2019023204A (en) 2019-02-14
JP2016179996A (en) 2016-10-13
TWI693224B (en) 2020-05-11
EP2822954B1 (en) 2016-03-09
PH12016500389A1 (en) 2018-10-01
MD20150064A2 (en) 2015-11-30
JP6411409B2 (en) 2018-10-24
ES2753548T3 (en) 2020-04-13
IL274988A (en) 2020-07-30
AU2023286038A1 (en) 2024-01-25
AU2021221427B2 (en) 2023-10-12
TW202104229A (en) 2021-02-01
PL3608325T3 (en) 2022-11-07
KR20220011789A (en) 2022-01-28
BR112015014714B1 (en) 2018-12-26
KR102406288B1 (en) 2022-06-13
PL3067358T3 (en) 2020-02-28
LTC2822954I2 (en) 2019-10-10
KR20210049956A (en) 2021-05-06
NL300947I2 (en) 2018-09-26
IL245780B (en) 2019-02-28
AU2016262722A1 (en) 2016-12-08
KR20230061576A (en) 2023-05-08
ZA201507997B (en) 2019-09-25
AU2020202914A1 (en) 2020-05-21
AU2013361401B2 (en) 2017-01-05
SG11201504857SA (en) 2015-07-30
MD4736C1 (en) 2021-07-31
KR101770048B1 (en) 2017-08-21
DK3608325T3 (en) 2022-08-22
HUE059688T2 (en) 2022-12-28
BR112015014714A2 (en) 2016-01-05
PT3608325T (en) 2022-10-17
AR094197A1 (en) 2015-07-15
AU2018232957B2 (en) 2020-01-30
MX2022013596A (en) 2022-11-16
EP3608325A1 (en) 2020-02-12
KR102351912B1 (en) 2022-01-17
NZ709260A (en) 2016-07-29
TW202212342A (en) 2022-04-01
CY2018022I2 (en) 2019-07-10
SI2822954T1 (en) 2016-07-29
UY35213A (en) 2014-06-30
EP3067358B1 (en) 2019-08-07
KR20220080208A (en) 2022-06-14
MY191741A (en) 2022-07-13
TWI642669B (en) 2018-12-01
BR122015029881B1 (en) 2022-04-26
US9216996B2 (en) 2015-12-22
US20170260204A1 (en) 2017-09-14
ZA201903516B (en) 2020-01-29
US9732092B2 (en) 2017-08-15
MY164352A (en) 2017-12-15
KR20200067930A (en) 2020-06-12
US20190092787A1 (en) 2019-03-28
CN111303152A (en) 2020-06-19
PH12015501445B1 (en) 2015-09-14
CR20150380A (en) 2015-08-21
HUE028284T2 (en) 2016-12-28
JP6028105B2 (en) 2016-11-16
SI3608325T1 (en) 2022-09-30
AU2020202914B2 (en) 2021-09-09
CN111303152B (en) 2023-04-11
DK2822954T3 (en) 2016-05-30
PT2822954E (en) 2016-06-16
AP2015008510A0 (en) 2015-06-30
CL2017001191A1 (en) 2018-02-09
US11548901B2 (en) 2023-01-10
KR102120875B1 (en) 2020-06-09
JP7301918B2 (en) 2023-07-03
MD4754C1 (en) 2021-12-31
MD4754B1 (en) 2021-05-31
TWI830624B (en) 2024-01-21
HRP20160544T1 (en) 2016-07-15
UA123572C2 (en) 2021-04-28
JP2016508134A (en) 2016-03-17
TWI815245B (en) 2023-09-11
PH12019501848A1 (en) 2020-03-02
PT3067358T (en) 2019-11-18
CA3131094A1 (en) 2014-06-26
MD4736B1 (en) 2020-12-31
IL239316A (en) 2016-09-29
CA2893843A1 (en) 2014-06-26
TW201441224A (en) 2014-11-01
SMT201600157B (en) 2016-08-31
EP3067358A1 (en) 2016-09-14
CA2893843C (en) 2018-09-04
US20210053988A1 (en) 2021-02-25
CA3012242C (en) 2021-11-02
HRP20191957T1 (en) 2020-02-07
AU2013361401A1 (en) 2015-08-27
EA030003B1 (en) 2018-06-29
AU2016262722B2 (en) 2018-10-18
KR102527797B1 (en) 2023-05-03
KR20190124814A (en) 2019-11-05
CN104995198B (en) 2018-01-30
KR20170073733A (en) 2017-06-28
US9663528B2 (en) 2017-05-30
EA201890236A1 (en) 2018-10-31
TWI752457B (en) 2022-01-11
AU2013361401C1 (en) 2018-08-09
ES2577283T3 (en) 2016-07-14
HK1250711A1 (en) 2019-01-11
MX357940B (en) 2018-07-31
US20140221356A1 (en) 2014-08-07
BR122015029881A2 (en) 2016-03-22
PE20151499A1 (en) 2015-10-29
JP2021193090A (en) 2021-12-23
PL2822954T3 (en) 2016-09-30
EP3608325B1 (en) 2022-07-06
US20160176885A1 (en) 2016-06-23
RS54873B1 (en) 2016-10-31
HUS1800035I1 (en) 2018-09-28
CY1122246T1 (en) 2020-11-25
JP2023134483A (en) 2023-09-27
KR20150096504A (en) 2015-08-24
DK3067358T3 (en) 2019-11-04
HK1205124A1 (en) 2015-12-11
PH12016500389B1 (en) 2018-10-01
SI3067358T1 (en) 2019-12-31
US20170057976A1 (en) 2017-03-02
CA3012242A1 (en) 2014-06-26
US10035809B2 (en) 2018-07-31
CL2015001756A1 (en) 2015-08-07
JP6571256B2 (en) 2019-09-04
JP2019218368A (en) 2019-12-26
MX344879B (en) 2017-01-11
HUE046559T2 (en) 2020-03-30
EA201591027A1 (en) 2016-05-31
CL2016000837A1 (en) 2016-10-21
LT3067358T (en) 2019-11-25
CN107674086B (en) 2020-03-31
PE20170528A1 (en) 2017-05-28
EP2822954A1 (en) 2015-01-14
HK1216643A1 (en) 2016-11-25
HRP20220899T1 (en) 2022-10-28

Similar Documents

Publication Publication Date Title
JP7301918B2 (en) Polycyclic carbamoylpyridone compound and pharmaceutical use thereof
OA17631A (en) Polycyclic-carbamoylpyridone compounds and their pharmaceutical use.

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13815937

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2013815937

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2893843

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 239316

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2015549669

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 001036-2015

Country of ref document: PE

Ref document number: MX/A/2015/008009

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12015501445

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 201591027

Country of ref document: EA

Ref document number: IDP00201503852

Country of ref document: ID

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015014714

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20200093

Country of ref document: MD

Kind code of ref document: A

Ref document number: 20150064

Country of ref document: MD

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: A 2015 0064

Country of ref document: MD

ENP Entry into the national phase

Ref document number: 20157019194

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15165942

Country of ref document: CO

Ref document number: 15265717

Country of ref document: CO

Ref document number: CR2015-000380

Country of ref document: CR

ENP Entry into the national phase

Ref document number: 2013361401

Country of ref document: AU

Date of ref document: 20131219

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112015014714

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150619

WWE Wipo information: entry into national phase

Ref document number: 12016500389

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 245780

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 000694-2016

Country of ref document: PE

WWE Wipo information: entry into national phase

Ref document number: P-2016/0407

Country of ref document: RS

WWE Wipo information: entry into national phase

Ref document number: 247311

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2017279

Country of ref document: CR

Ref document number: CR2017-000279

Country of ref document: CR